WO2014189973A2 - Anti-transferrin receptor antibodies and methods of use - Google Patents

Anti-transferrin receptor antibodies and methods of use Download PDF

Info

Publication number
WO2014189973A2
WO2014189973A2 PCT/US2014/038847 US2014038847W WO2014189973A2 WO 2014189973 A2 WO2014189973 A2 WO 2014189973A2 US 2014038847 W US2014038847 W US 2014038847W WO 2014189973 A2 WO2014189973 A2 WO 2014189973A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
hvr
nos
amino acid
Prior art date
Application number
PCT/US2014/038847
Other languages
English (en)
French (fr)
Other versions
WO2014189973A3 (en
Inventor
Yin Zhang
Joy Yu Zuchero
Jasvinder Atwal
Jessica COUCH
Mark Dennis
James Ernst
Ryan Watts
Gregory A. Lazar
Original Assignee
Genentech, Inc.
F. Hoffmann-La Roche Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to BR112015029009-4A priority Critical patent/BR112015029009B1/pt
Priority to EP19190228.7A priority patent/EP3594240B1/en
Application filed by Genentech, Inc., F. Hoffmann-La Roche Ag filed Critical Genentech, Inc.
Priority to PE2020002132A priority patent/PE20210649A1/es
Priority to EP23210472.9A priority patent/EP4324480A3/en
Priority to IL242088A priority patent/IL242088B2/en
Priority to EA201592213A priority patent/EA038367B1/ru
Priority to MA38632A priority patent/MA38632B1/fr
Priority to CN201480035703.6A priority patent/CN105358578B/zh
Priority to AU2014268726A priority patent/AU2014268726B2/en
Priority to EP14732721.7A priority patent/EP2999716A2/en
Priority to KR1020157035600A priority patent/KR102293064B1/ko
Priority to CA2908743A priority patent/CA2908743A1/en
Priority to JP2016515030A priority patent/JP6612214B2/ja
Priority to MX2015015970A priority patent/MX2015015970A/es
Priority to SG11201509566RA priority patent/SG11201509566RA/en
Priority to UAA201512536A priority patent/UA119235C2/uk
Publication of WO2014189973A2 publication Critical patent/WO2014189973A2/en
Publication of WO2014189973A3 publication Critical patent/WO2014189973A3/en
Priority to PH12015502565A priority patent/PH12015502565B1/en
Priority to CR20150618A priority patent/CR20150618A/es
Priority to HK16103512.5A priority patent/HK1215584A1/zh
Priority to AU2020200355A priority patent/AU2020200355A1/en
Priority to AU2022201773A priority patent/AU2022201773A1/en
Priority to IL304243A priority patent/IL304243A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2881Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD71
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/71Decreased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/72Increased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present invention relates to anti-transferrin receptor antibodies and methods of using the same.
  • BBB blood-brain barrier
  • BBB blood-brain barrier
  • the anti-BBB receptor antibody can be made multispecific to target one or more desired antigens in the CNS, or one or more heterologous molecules can be coupled to the anti- BBB receptor antibody; in either case, the anti-BBB receptor antibody can assist in delivering a therapeutic molecule into the CNS across the BBB.
  • BBB-R high affinity anti-BBB receptor
  • anti-TfR A from Atwal et al. and Yu et al., supra
  • BBB-R high affinity anti-BBB receptor antibodies
  • lowering affinity for the BBB-R improves brain uptake and distribution, with a robust shift observed in localization from the vasculature to neurons and associated neuropil distributed within the CNS.
  • the lower affinity of the antibody for the BBB-R is proposed to impair the ability of the antibody to return to the vascular side of the BBB via the BBB-R from the CNS side of the membrane because the overall affinity of the antibody for the BBB-R is low and the local concentration of the antibody on the CNS side of the BBB is non-saturating due to the rapid dispersal of the antibody into the CNS compartment.
  • the circulating antibody levels of the lower-affinity antibody are sustained at therapeutic levels for a longer period of time than the higher-affinity antibody, which consequently improves uptake of antibody in brain for a longer period of time. Furthermore, this improvement in both plasma and brain exposure may reduce the frequency of dosing in the clinic, which would have potential benefit not only for patient compliance and convenience but also in lessening any potential side effects or off-target effects of the antibody and/or of a therapeutic compound coupled thereto.
  • the low-affinity BBB-R antibodies described in the above-referenced work were selected /engineered to avoid interference with the natural binding between transferrin and the TfR, and thus to avoid potential iron transport-related side effects. Nonetheless, upon administration of certain of these antibodies in mice, some marked side effects were observed.
  • the mice displayed a primary response of robust depletion of reticulocyte populations accompanied by rapid onset acute clinical symptoms. Though the mice recovered from both the acute clinical symptoms and the decreased reticulocyte levels in due course, avoiding or otherwise mitigating this impact on reticulocytes is clearly desirable for an anti-TfR antibody to be able to be used safely as a therapeutic molecule.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • the invention provides antibodies and functional parts thereof which do specifically recognize both primate and human TfR, in order to facilitate safety and efficacy studies in primates with the antibodies prior to therapeutic or diagnostic use in humans.
  • In vitro studies using a human erythroblast cell line and primary bone marrow cells treated with the anti-human TfR antibodies of the invention demonstrated that a robust depletion of TfR-positive erythroid cells is also observable in human/primate cellular systems as it is in mice (see, e.g., Example 4).
  • modifications to the antibodies of the invention to greatly reduce or eliminate the unwanted reduction in the Tf -expressing reticulocyte population upon anti-TfR administration while still enabling the enhanced BBB transport, increased CNS distribution and CNS retention provided by the anti-human/primate Tf antibodies administered at therapeutic concentrations.
  • Several general approaches to mitigate the observed effect of the anti-TfR antibodies of the invention on both the primary and residual reticulocyte depletion are provided herein, and may be used singly or in combination.
  • the effector function of the anti-human/cyno TfR antibody is reduced or eliminated in order to reduce or eliminate ADCC activity.
  • the affinity of the anti-human/cyno TfR antibody for human or primate TfR is further lessened such that interactions of the antibody with the reticulocyte population are less detrimental to that population.
  • a third approach is directed to reducing the amount of anti-human/cyno TfR antibody that is present in the plasma to reduce exposure of the reticulocyte population to potentially detrimental concentrations of the antibody.
  • a fourth approach seeks to protect, stabilize and/or replenish reticulocyte populations such that any potential depletion of the reticulocyte population in circulation or in bone marrow by administration of the anti- human/cyno TfR antibody is avoided, lessened, or mitigated.
  • Effector function reduction or elimination may be accomplished by: (i) reduction or elimination of wild-type mammalian glycosylation of the antibody, (for example, by producing the antibody in an environment where such glycosylation cannot occur, by mutating one or more carbohydrate attachment points such that the antibody cannot be glycosylated, or by chemically or enzymatically removing one or more carbohydrates from the antibody after it has been glycosylated); (ii) by reduction or elimination of the Fc receptor- binding capability of the anti-human/cyno TfR antibody (for example, by mutation of the Fc region, by deletion within the Fc region or elimination of the Fc region); or (iii) by utilization of an antibody isotype known to have minimal or no effector function (ie., including but not limited to IgG4).
  • Decreasing antibody complement activation may be accomplished by reduction or elimination of the Clq binding capability of the anti-human/cyno TfR antibody (for example, by mutation of, deletion within or elimination of the Fc region, or by modifying the non-Fc portion of the anti-human/cyno TfR antibody), or by otherwise suppressing activation or activity of the complement system (for example, by co-administering one or more complement pathway activation or complement pathway activity inhibitors).
  • Reducing the amount of anti-human/cyno TfR antibody present in the plasma in order to reduce exposure of the reticulocyte population to potentially detrimental concentrations of the antibody may be accomplished in several ways.
  • One method is to simply decrease the amount of the antibody that is dosed, potentially while also increasing the frequency of the dosing, such that the maximal concentration in the plasma is lowered but a sufficient serum level is maintained for efficacy, while still below the threshold of the cell-depleting side effect.
  • Another method which may be combined with dosing modifications, is to select or engineer an anti-TfR antibody that has pH-sensitive binding to TfR such that it binds to cell surface TfR in the plasma at pH 7.4 with desirably low affinity as described herein, but upon internalization into an endosomal compartment, such binding to TfR is rapidly and significantly reduced at the relatively lower pH of that compartment (pH 5.5-6.0).
  • Such dissociation may protect the antibody from antigen-mediated clearance, or increase the amount of antibody that is either delivered to the CNS or recycled back across the BBB - in either case, the effective concentration of the antibody is increased relative to an anti-TfR antibody that does not comprise such pH sensitivity, without increasing the administered dose of the antibody, and in turn potentially permitting a lower dose of the antibody with concomitantly lesser risk of side effects.
  • Protecting, stabilizing and/or replenishing reticulocyte populations may be
  • At least one further therapeutic agent may be coadministered (simultaneously or sequentially) that mitigates negative side effects of the antibody on reticulocyte populations.
  • therapeutic agents include, but are not limited to, erythropoietin (EPO), iron supplements, vitamin C, folic acid, and vitamin B12.
  • EPO erythropoietin
  • iron supplements iron supplements
  • vitamin C folic acid
  • vitamin B12 vitamin B12.
  • Physical replacement of red blood cells ie, reticulocytes
  • transfusion with similar cells which may be from another individual of similar blood type or may have been previously extracted from the subject to whom the anti-human cyno TfR antibody is administered.
  • any combination of the foregoing methods may be employed to engineer an antibody (and/or dosage regimen for same) with the optimum balance between (i) the desirably low affinity for primate or human TfR that will maximize transport of the antibody and any conjugated compounds into the CNS; (ii) the affinity of the conjugated compound (including as a nonlimiting example, a second or further antigen-binding specificity in the anti-human/cyno TfR antibody) for its CNS antigen, since this is relevant to the amount of the compound that needs to be present in the CNS to have a therapeutic effect; (iii) the clearance rate of the anti-human/cynoTfR antibody; (iv) the lability of the anti-TfR/conjugated compound at low pH to facilitate release of the conjugated compound on the CNS/brain side of the BBB, and (v) the impact on reticulocyte populations.
  • the affinity of the conjugated compound including as a nonlimiting example, a second or further antigen-binding specific
  • the reticulocyte-depleting effect recognized herein of anti-TfR antibody administration may be useful in the treatment of any disease or disorder where overproliferation of reticulocytes is problematic.
  • reticulocytes in congenital polycythemia or neoplastic polycythemia vera, raised red blood cell counts due to hyperproliferation of, e.g., reticulocytes, results in thickening of blood and concomitant physiological symptoms.
  • Administration of an anti-human/cyno TfR antibody of the invention wherein at least partial effector function of the antibody was preserved would permit selective removal of immature reticulocyte populations without impacting normal transferrin transport into the CNS. Dosing of such an antibody could be modulated such that acute clinical symptoms could be minimized (ie, by dosing at a very low dose or at widely-spaced intervals), as well-understood in the art.
  • Anti-TfR/BACEl and anti-TfR/ Abeta are each promising and novel therapeutic candidates for the treatment of Alzheimer's disease.
  • receptor mediated transport (RMT)-based bispecific targeting technology opens the door for a wide range of potential therapeutics for CNS diseases.
  • the invention provides methods of engineering BBB-penetrant therapeutics that greatly improve transport across the BBB and CNS distribution of the therapeutic without depletion of reticulocytes.
  • the invention provides an isolated antibody that binds to human transferrin receptor (TfR) and primate TfR, wherein the antibody does not inhibit the binding of transferrin to TfR.
  • the binding is specific binding.
  • the antibody further does not inhibit the binding of human hemachromatosis protein ("HFE") to TfR.
  • HFE human hemachromatosis protein
  • the binding is specific binding.
  • the antibody is a monoclonal antibody.
  • the antibody is a human antibody.
  • the antibody is a humanized antibody.
  • the antibody is a chimeric antibody.
  • the antibody is an antibody fragment that binds human TfR and primate TfR.
  • the primate TfR is from cynomolgous monkey.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 31, 33 and 34. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 36, 38 and 39. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 36, 40 and 34. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 42, 43 and 44.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 31, 33 and 34. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 46, 48 and 49. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 52, 54 and 55.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 52, 58 and 59. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 62, 63 and 55. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 52, 65 and 55.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 68, 69 and 70. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 73, 75 and 76. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 79, 81 and 82.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 79, 83 and 84. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 87, 89 and 90. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 93, 95 and 96.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 99, 101 and 102. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 127, 33 and 34. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 52, 156 and 55.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 52, 157 and 55. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 155, 54 and 55
  • the antibody comprises HVR-H1 , HVR-H2 and HVR-H3, respectively comprising the amino acid sequences of SEQ ID NOs: 32, 33 and 34.
  • the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 37, 38 and 39.
  • the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 32, 40 and 34.
  • the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 37, 43 and 44. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 32, 33 and 34. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 47, 48 and 49.
  • the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 54 and 55. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 58 and 59. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 63 and 55.
  • the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 65 and 55. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 69 and 70. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 74, 75 and 76.
  • the antibody comprises HVR-H1, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 80, 81 and 82. In another aspect of the above embodiment, the antibody comprises HVR-H1, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 80, 83 and 84. In another aspect of the above embodiment, the antibody comprises HVR-H1, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 88, 89 and 90.
  • the antibody comprises HVR-H1, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 94, 95 and 96. In another aspect of the above embodiment, the antibody comprises HVR-H1, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 100, 101 and 102. In another aspect of the above embodiment, the antibody comprises HVR-H1, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 156 and 55. In another aspect of the above embodiment, the antibody comprises HVR-H1, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 157 and 55.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 31. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 35, 30 and 36. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 41, 30 and 42. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 31.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 45, 30 and 46. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 50, 51 and 52. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 56, 57 and 52.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 60, 61 and 62. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 60, 64 and 52. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 66, 67 and 68.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 71, 72 and 73. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs:77, 78 and 79. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 85, 86 and 87.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 91, 92 and 93. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 97, 98 and 99. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 127. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 50, 51 and 155.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 31 and HVR-Hl, HVR-H2 and HVR-H3, respectively comprising the amino acid sequences of SEQ ID NOs: 32, 33 and 34.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 35, 30 and 36 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 37, 38 and 39.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 35, 30 and 36 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 32, 40 and 34.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 41, 30 and 42 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 37, 43 and 44
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 31 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 32, 33 and 34.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 45, 30 and 46 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 47, 48 and 49.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 50, 51 and 52 and HVR-Hl , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 54 and 55.
  • the antibody comprises HVR-L1 , HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 56, 57 and 52 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 58 and 59.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 60, 61 and 62 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 63 and 55.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 60, 64 and 52 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 65 and 55.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 66, 67 and 68 and HVR-Hl , HVR-H2, and HVR- H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 69 and 70.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 71 , 72 and 73 and HVR-Hl , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 74, 75 and 76.
  • the antibody comprises HVR-Ll , HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs:77, 78 and 79 and HVR-Hl , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 80, 81 and 82.
  • the antibody comprises HVR-Ll, HVR-L2 and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 77, 78 and 79, and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 80, 83 and 84.
  • the antibody comprises HVR-Ll , HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 85, 86 and 87 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 88, 89 and 90.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 91, 92 and 93 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 94, 95 and 96.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 97, 98 and 99 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 100, 101 and 102.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 127 and HVR-Hl, HVR-H2 and HVR- H3 respectively comprising the amino acid sequences of SEQ ID NOs: 32, 33 and 34.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 50, 51 and 52 and HVR-Hl, HVR-H2 and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 156 and 55.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 50, 51 and 52 and HVR-Hl, HVR-H2 and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 157 and 55.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 50, 51 and 155 and HVR-Hl, HVR-H2 and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 54 and 55.
  • the antibody comprises at least one HVR-Hl, HVR-H2, HVR-H3, HVR-Ll, HVR-L2, or HVR-L3 sequence selected from the group of sequences consisting of an HVR-Hl sequence of SEQ ID NO: 47, 53, or 100; an HVR-H2 sequence of SEQ ID NO: 48, 69, 101, 156 or 157;an HVR-H3 sequence of SEQ ID NO: 49, 76, or 102; an HVR-Ll sequence of SEQ ID NO: 45, 66 or 97; an HVR-L2 sequence of SEQ ID NOs: 30, 67 or 98; and an HVR-L3 sequence of SEQ ID NOs: 46, 68 or 102.
  • the antibody comprises (a) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 7, 8, 9, 10, 15, 16, 17, 18, 20, 25, 26, 27, 28, 108, 114, 120, 126, 153 or 154; (b) a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:4, 5, 6, 11, 12, 13, 14, 19, 21, 22, 23, 24, 105, 111, 117, 123 or 151; or (c) a VH sequence as in (a) and a VL sequence as in (b).
  • the antibody comprises a VL sequence of SEQ ID NO: 4 and a VH sequence of SEQ ID NO: 7.
  • the antibody comprises a VL sequence of SEQ ID NO: 5 and a VH sequence of SEQ ID NO: 8. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 5 and a VH sequence of SEQ ID NO: 9. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 6 and a VH sequence of SEQ ID NO: 10. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 11 and a VH sequence of SEQ ID NO: 15. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 12 and a VH sequence of SEQ ID NO: 16.
  • the antibody comprises a VL sequence of SEQ ID NO: 13 and a VH sequence of SEQ ID NO: 17. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 14 and a VH sequence of SEQ ID NO: 18. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 19 and a VH sequence of SEQ ID NO: 20. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 21 and a VH sequence of SEQ ID NO: 25. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 22 and a VH sequence of SEQ ID NO: 26.
  • the antibody comprises a VL sequence of SEQ ID NO: 23 and a VH sequence of SEQ ID NO: 27. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 24 and a VH sequence of SEQ ID NO: 28. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 105 and a VH sequence of SEQ ID NO: 108. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 111 and a VH sequence of SEQ ID NO: 114. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 1 17 and a VH sequence of SEQ ID NO: 120.
  • the antibody comprises a VL sequence of SEQ ID NO: 123 and a VH sequence of SEQ ID NO: 126. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 105 and a VH sequence of SEQ ID NO: 153. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 105 and a VH sequence of SEQ ID NO: 154. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 151 and a VH sequence of SEQ ID NO: 108.
  • the antibody comprises a VH sequence of (a) SEQ ID NO: 108, (b) SEQ ID NO: 114, (c) SEQ ID NO: 120 or (d) SEQ ID NO: 126.
  • the antibody comprises a VL sequence of (a) SEQ ID NO: 105, (b) SEQ ID NO: 111, (c) SEQ ID NO: 1 17, or (d) SEQ ID NO: 123.
  • the antibody is selected from the group consisting of antibodies 7A4, 8A2, 15D2, 10D1 1 , 7B10, 15G11, 16G5, 13C3, 16G4, 16F6, 7G7, 4C2, 1B12, and 13D4.
  • the antibody is 7A4.
  • the antibody is 8A2.
  • the antibody is 15D2.
  • the antibody is 10D11.
  • the antibody is 7B10.
  • the antibody is 15G11.
  • the antibody is 16G5.
  • the antibody is 13C3.
  • the antibody is 16G4.
  • the antibody is 16F6.
  • the antibody is 7G7.
  • the antibody is 4C2. In another such aspect, the antibody is IB 12. In another such aspect, the antibody is 13D4. In one aspect of any of the foregoing, the antibody is further affinity matured. In one such aspect, the antibody is selected from the group consisting of 15Gl l .vl , 15Gl l .v2, 15G11.V3, 15G11.V4, 15Gl l.v5, 7A4.vl; 7A4.v2, 7A4.v3, 7A4.v4, 7A4.v5, 7A4.v6, 7A4.v7, 7A4.v8, 7A4.v9, 7A4.vlO, 7A4.vl l, 7A4.vl2, 7A4.vl3, 7A4.vl4, 7A4.vl5, 7G7.vl, 16F6.vl, 16F6.v2, 16F6.v3, 16F6.v4, 15G11.N52A, 15G11.T53A and 15G11.W92A.
  • the antibody is 15Gl l.vl . In another such aspect, the antibody is 15Gl l.v2. In another such aspect, the antibody is 15Gl l .v3. In another such aspect, the antibody is 15Gl l .v4. In another such aspect, the antibody is 15G1 l.v5. In another such aspect, the antibody is 7A4.vl . In another such aspect, the antibody is 7A4.v2. In another such aspect, the antibody is 7A4.v3. In another such aspect, the antibody is 7A4.v4. In another such aspect, the antibody is 7A4.v5. In another such aspect, the antibody is 7A4.v6. In another such aspect, the antibody is 7A4.v7.
  • the antibody is 7A4.v8. In another such aspect, the antibody is 7A4.v9. In another such aspect, the antibody is 7A4.vlO. In another such aspect, the antibody is 7A4.vl l. In another such aspect, the antibody is 7A4.vl2. In another such aspect, the antibody is 7A4.vl 3. In another such aspect, the antibody is 7A4.vl4. In another such aspect, the antibody is 7A4.vl5. In another such aspect, the antibody is 7G7.vl. In another such aspect, the antibody is 16F6.vl . In another such aspect, the antibody is 16F6.v2. In another such aspect, the antibody is 16F6.v3. In another such aspect, the antibody is 16F6.v4. In another such aspect, the antibody is 15G11.N52A. In another such aspect, the antibody is 15G11.T53A. In another such aspect, the antibody is 15G11.W92A.
  • the antibody is modified at one or more amino acid positions in the VH or VL to the amino acid indicated for that position in Figures 4E-1 and 4E-2.
  • the antibody comprises a sequence or one or more HVR sequences corresponding to that of or one or more of those set forth for any one of the clones in Figures 3 and 4 and Figures 4E-1 and 4E-2.
  • the antibody comprises a VH or VL sequence corresponding to that of set forth for any one of the clones in Figures 3 and 4 and Figures 4E-1 and 4E-2.
  • the antibody comprises one or more HVR sequences corresponding to one or more of those set forth for any one of the clones in Figures 3 and 4 and Figures 4E- 1 and 4E-2.
  • the antibody is coupled to a therapeutic compound. In another aspect of the above embodiment, the antibody is coupled to an imaging agent or a label. In one such aspect, the antibody is a multispecific antibody and the therapeutic compound optionally forms one portion of the multispecific antibody. In one such aspect, the multispecific antibody comprises a first antigen binding site which binds TfR and a second antigen binding site which binds a brain antigen.
  • the brain antigen is selected from the group consisting of: beta-secretase 1 (BACE1), Abeta, epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), tau, apolipoprotein E (ApoE), alpha-synuclein, CD20, huntingtin, prion protein (PrP), leucine rich repeat kinase 2 (LRRK2), parkin, presenilin 1, presenilin 2, gamma secretase, death receptor 6 (DR6), amyloid precursor protein (APP), p75 neurotrophin receptor (p75NTR), and caspase 6.
  • BACE1 beta-secretase 1
  • Abeta Abeta
  • EGFR epidermal growth factor receptor
  • HER2 human epidermal growth factor receptor 2
  • tau tau
  • alpha-synuclein CD20
  • huntingtin huntingtin
  • prion protein PrP
  • the invention provides an isolated nucleic acid encoding any of the foregoing antibodies.
  • the invention provides a host cell comprising such nucleic acid.
  • the invention provides a method of producing any of the foregoing antibodies comprising culturing such host cell so that the antibody is produced and optionally further comprising recovering the antibody from the host cell.
  • the invention provides a pharmaceutical formulation comprising any of the foregoing antibodies and a pharmaceutically acceptable carrier.
  • the invention provides any of the foregoing antibodies for use as a medicament. In another aspect of the above embodiment, the invention provides the use of any of the foregoing antibodies in the manufacture of a medicament for treating a neurological disorder.
  • the neurological disorder is selected from the group consisting of a neuropathy disorder, a neurodegenerative disease, cancer, an ocular disease disorder, a seizure disorder, a lysosomal storage disease, amyloidosis, a viral or microbial disease, ischemia, a behavioral disorder, and CNS inflammation.
  • the invention provides any of the foregoing antibodies for use in treating a neurological disorder.
  • the neurological disorder is selected from the group consisting of a neuropathy disorder, a neurodegenerative disease, cancer, an ocular disease disorder, a seizure disorder, a lysosomal storage disease, amyloidosis, a viral or microbial disease, ischemia, a behavioral disorder, and CNS inflammation.
  • the invention provides any of the foregoing antibodies for use in transporting one or more compounds across the BBB.
  • use of any of the foregoing antibodies in the manufacture of a medicament for transporting one or more compounds across the BBB is provided.
  • a method of transporting a compound across the BBB in a subject comprising exposing any of the foregoing antibodies to the BBB such that the antibody transports the compound coupled thereto across the BBB.
  • the BBB is in a human subject.
  • the dose amount and/or frequency of administration is modulated to reduce the concentration of antibody to which red blood cells are exposed.
  • the method further comprises the step of monitoring the subject for depletion of red blood cells.
  • the antibody coupled to the compound is administered at a therapeutic dose.
  • the therapeutic dose is TfR-saturating.
  • administration of the antibody is at a dose and/or dose frequency calibrated to minimize acute clinical symptoms of the antibody administration.
  • a method of increasing exposure of the CNS of a subject to a compound comprising exposing any of the foregoing antibodies to the BBB such that the antibody transports the compound coupled thereto across the BBB.
  • the BBB is in a human subject.
  • the dose amount and/or frequency of administration is modulated to reduce the concentration of antibody to which red blood cells are exposed.
  • the method further comprises the step of monitoring the subject for depletion of red blood cells.
  • the antibody coupled to the compound is administered at a therapeutic dose.
  • the therapeutic dose is TfR-saturating.
  • administration of the antibody is at a dose and/or dose frequency calibrated to minimize acute clinical symptoms of the antibody administration.
  • a method of increasing rentention in the CNS of a compound administered to a subject comprising exposing any of the foregoing antibodies to the BBB such that the retention in the CNS of the compound is increased.
  • the BBB is in a human subject.
  • the dose amount and/or frequency of administration is modulated to reduce the concentration of antibody to which red blood cells are exposed.
  • the method further comprises the step of monitoring the subject for depletion of red blood cells.
  • the antibody coupled to the compound is administered at a therapeutic dose.
  • the therapeutic dose is TfR-saturating.
  • administration of the antibody is at a dose and/or dose frequency calibrated to minimize acute clinical symptoms of the antibody administration.
  • a method of treating a neurological disorder in a mammal comprising treating the mammal with any of the foregoing antibodies.
  • the neurological disorder is selected from the group consisting of a neuropathy disorder, a neurodegenerative disease, cancer, an ocular disease disorder, a seizure disorder, a lysosomal storage disease, amyloidosis, a viral or microbial disease, ischemia, a behavioral disorder, and CNS inflammation.
  • the neurological disorder is in a human subject.
  • the dose amount and/or frequency of administration is modulated to reduce the concentration of antibody to which the re blood cells are exposed.
  • the method further comprises the step of monitoring the subject for depletion of red blood cells.
  • the antibody coupled to the compound is administered at a therapeutic dose.
  • the therapeutic dose is TfR-saturating.
  • administration of the antibody is at a dose and/or dose frequency calibrated to minimize acute clinical symptoms of the antibody administration.
  • the invention provides an isolated antibody that binds to human TfR and primate TfR, wherein the antibody does not inhibit the binding of transferrin to TfR, and wherein one or more properties of the antibody have been modified to reduce or eliminate the impact of the antibody on reticulocytes and/or reduce the severity or presence of acute clinical symptoms in a subject or mammal treated with the antibody.
  • the binding is specific binding.
  • the antibody further does not inhibit the binding of HFE to TfR.
  • the antibody is a monoclonal antibody.
  • the antibody is a human antibody.
  • the antibody is a humanized antibody.
  • the antibody is a chimeric antibody.
  • the antibody is an antibody fragment that binds human TfR and primate TfR.
  • the primate TfR is from cynomolgous monkey.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 31, 33 and 34. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 36, 38 and 39. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 36, 40 and 34. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 42, 43 and 44.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 31, 33 and 34. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 46, 48 and 49. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs:52, 54 and 55.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 52, 58 and 59. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 62, 63 and 55. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs:52, 65 and 55.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 68, 69 and 70. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 73, 75 and 76. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 79, 81 and 82.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 79, 83 and 84. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs:87, 89 and 90. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 93, 95 and 96.
  • the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 99, 101 and 102. In another aspect of the above embodiment, the antibody comprises HVR-L3, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 127, 33 and 34.
  • the antibody comprises HVR-Hl, HVR-H2 and HVR-H3, respectively comprising the amino acid sequences of SEQ ID NOs: 32, 33 and 34.
  • the antibody comprises HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 37, 38 and 39.
  • the antibody comprises HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 32, 40 and 34.
  • the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 37, 43 and 44. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 32, 33 and 34. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 47, 48 and 49.
  • the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 54 and 55. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 58 and 59. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 63 and 55.
  • the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 65 and 55. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 3, 69 and 70. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 74, 75 and 76.
  • the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 80, 81 and 82. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 80, 83 and 84. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 88, 89 and 90.
  • the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 94, 95 and 96. In another aspect of the above embodiment, the antibody comprises HVR-H1 , HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 100, 101 and 102.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 31. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 35, 30 and 36. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 41 , 30 and 42. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 31.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 45, 30 and 46. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 50, 51 and 52. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 56, 57 and 52.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 60, 61 and 62. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 60, 64 and 52. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 66, 67 and 68.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 71, 72 and 73. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs:77, 78 and 79. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 85, 86 and 87.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 91, 92 and 93. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 97, 98 and 99. In another aspect of the above embodiment, the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 127.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 31 and HVR-H1, HVR-H2 and HVR-H3, respectively comprising the amino acid sequences of SEQ ID NOs: 32, 33 and 34.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 35, 30 and 36 and HVR-H1, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 37, 38 and 39.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 35, 30 and 36 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 32, 40 and 34.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 41, 30 and 42 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 37, 43 and 44
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 31 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 32, 33 and 34.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 45, 30 and 46 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 47, 48 and 49.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 50, 51 and 52 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 54 and 55.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 56, 57 and 52 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 58 and 59.
  • the antibody comprises HVR-L1, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 60, 61 and 62 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 63 and 55.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 60, 64 and 52 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 65 and 55.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 66, 67 and 68 and HVR-Hl, HVR-H2, and HVR- H3 respectively comprising the amino acid sequences of SEQ ID NOs: 53, 69 and 70.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 71, 72 and 73 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 74, 75 and 76.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs:77, 78 and 79 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 80, 81 and 82.
  • the antibody comprises HVR-Ll, HVR-L2 and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 77, 78 and 79, and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 80, 83 and 84.
  • the antibody comprises HVR-Ll , HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 85, 86 and 87 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 88, 89 and 90.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 91, 92 and 93 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 94, 95 and 96.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 97, 98 and 99 and HVR-Hl, HVR-H2, and HVR-H3 respectively comprising the amino acid sequences of SEQ ID NOs: 100, 101 and 102.
  • the antibody comprises HVR-Ll, HVR-L2, and HVR-L3 respectively comprising the amino acid sequences of SEQ ID NOs: 29, 30 and 127 and HVR-Hl , HVR-H2 and HVR- H3 respectively comprising the amino acid sequences of SEQ ID NOs: 32, 33 and 34.
  • the antibody comprises at least one HVR-Hl , HVR- H2, HVR-H3, HVR-Ll, HVR-L2, or HVR-L3 sequence selected from the group of sequences consisting of an HVR-Hl sequence of SEQ ID NO: 47, 53, or 100; an HVR-H2 sequence of SEQ ID NO: 48, 69, or 101 ;an HVR-H3 sequence of SEQ ID NO: 49, 76, or 102; an HVR-Ll sequence of SEQ ID NO: 45, 66 or 97; an HVR-L2 sequence of SEQ ID NOs: 30, 67 or 98; and an HVR-L3 sequence of SEQ ID NOs: 46, 68 or 102.
  • the antibody comprises (a) a VH sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 7, 8, 9, 10, 15, 16, 17, 18, 20, 25, 26, 27, 28, 108, 114, 120 or 126; (b) a VL sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:4, 5, 6, 1 1 , 12, 13, 14, 19, 21 , 22, 23, 24, 105, 111 , 117 or 123; or (c) a VH sequence as in (a) and a VL sequence as in (b).
  • the antibody comprises a VL sequence of SEQ ID NO: 4 and a VH sequence of SEQ ID NO: 7.
  • the antibody comprises a VL sequence of SEQ ID NO: 5 and a VH sequence of SEQ ID NO: 8. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 5 and a VH sequence of SEQ ID NO: 9. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 6 and a VH sequence of SEQ ID NO: 10. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 1 1 and a VH sequence of SEQ ID NO: 15. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 12 and a VH sequence of SEQ ID NO: 16.
  • the antibody comprises a VL sequence of SEQ ID NO: 13 and a VH sequence of SEQ ID NO: 17. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 14 and a VH sequence of SEQ ID NO: 18. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 19 and a VH sequence of SEQ ID NO: 20. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 21 and a VH sequence of SEQ ID NO: 25. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 22 and a VH sequence of SEQ ID NO: 26.
  • the antibody comprises a VL sequence of SEQ ID NO: 23 and a VH sequence of SEQ ID NO: 27. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 24 and a VH sequence of SEQ ID NO: 28. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 105 and a VH sequence of SEQ ID NO: 108. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 111 and a VH sequence of SEQ ID NO: 114. In another such aspect, the antibody comprises a VL sequence of SEQ ID NO: 1 17 and a VH sequence of SEQ ID NO: 120.
  • the antibody comprises a VL sequence of SEQ ID NO: 123 and a VH sequence of SEQ ID NO: 126.
  • the antibody comprises a VH sequence of (a) SEQ ID NO: 108, (b) SEQ ID NO: 114, (c) SEQ ID NO: 120 or (d) SEQ ID NO: 126.
  • the antibody comprises a VL sequence of (a) SEQ ID NO: 105, (b) SEQ ID NO: 111, (c) SEQ ID NO: 1 17, or (d) SEQ ID NO: 123.
  • the antibody is selected from the group consisting of antibodies 7A4, 8A2, 15D2, 10D1 1 , 7B10, 15G11, 16G5, 13C3, 16G4, 16F6, 7G7, 4C2, 1B12, and 13D4.
  • the antibody is 7A4.
  • the antibody is 8A2.
  • the antibody is 15D2.
  • the antibody is 10D11.
  • the antibody is 7B10.
  • the antibody is 15G11.
  • the antibody is 16G5.
  • the antibody is 13C3.
  • the antibody is 16G4.
  • the antibody is 16F6.
  • the antibody is 7G7.
  • the antibody is 4C2.
  • the antibody is IB 12.
  • the antibody is 13D4.
  • the antibody is further affinity matured.
  • the antibody is selected from the group consisting of 15Gl l .vl , 15Gl l .v2, 15G11.V3, 15G1 1.V4, 15Gl l .v5, 7A4.vl; 7A4.v2, 7A4.v3, 7A4.v4, 7A4.v5, 7A4.v6, 7A4.v7, 7A4.v8, 7A4.v9, 7A4.vlO, 7A4.vl l, 7A4.vl2, 7A4.vl3, 7A4.vl4, 7A4.vl5, 7G7.vl , 16F6.vl , 16F6.v2, 16F6.v3 and 16F6.v4.
  • the antibody is 15G1 l .vl . In another such aspect, the antibody is 15G1 l .v2. In another such aspect, the antibody is 15G1 l .v3. In another such aspect, the antibody is 15Gl l .v4. In another such aspect, the antibody is 15Gl l .v5. In another such aspect, the antibody is 7A4.vl . In another such aspect, the antibody is 7A4.v2. In another such aspect, the antibody is 7A4.v3. In another such aspect, the antibody is 7A4.v4. In another such aspect, the antibody is 7A4.v5. In another such aspect, the antibody is 7A4.v6. In another such aspect, the antibody is 7A4.v7.
  • the antibody is 7A4.v8. In another such aspect, the antibody is 7A4.v9. In another such aspect, the antibody is 7A4.vlO. In another such aspect, the antibody is 7A4.vl l . In another such aspect, the antibody is 7A4.vl2. In another such aspect, the antibody is 7A4.vl3. In another such aspect, the antibody is 7A4.vl4. In another such aspect, the antibody is 7A4.vl5. In another such aspect, the antibody is 7G7.vl . In another such aspect, the antibody is 16F6.vl . In another such aspect, the antibody is 16F6.v2. In another such aspect, the antibody is 16F6.v3. In another such aspect, the antibody is 16F6.v4.
  • the antibody is 15G1 1.N52A. In another such aspect, the antibody is 16F6.v4. In another such aspect, the antibody is 15G11.T53A. In another such aspect, the antibody is 16F6.v4. In another such aspect, the antibody is 15G1 1.W92A. In another such aspect, the antibody is 16F6.v4.
  • the antibody is modified at one or more amino acid positions in the VH or VL to the amino acid indicated for that position in Figures 4E-1 and 4E-2.
  • the antibody comprises a sequence or one or more HVR sequences corresponding to that of or one or more of those set forth for any one of the clones in Figures 3 and 4 and Figures 4E-1 and 4E-2.
  • the antibody comprises a VH or VL sequence corresponding to that of set forth for any one of the clones in Figures 3 and 4 and Figures 4E-1 and 4E-2.
  • the antibody comprises one or more HVR sequences corresponding to one or more of those set forth for any one of the clones in Figures 3 and 4 and Figures 4E- 1 and 4E-2.
  • the one or more properties of the antibody are selected from the effector function of the antibody Fc region, the complement activation function of the antibody and the affinity of the antibody for TfR.
  • the property is the effector function of the antibody Fc region.
  • the property is the complement activation function of the antibody.
  • the property is the affinity of the antibody for TfR.
  • the effector function or complement activation function has been reduced or eliminated relative to a wild-type antibody of the same isotype.
  • the effector function is reduced or eliminated by a method selected from reduction of glycosylation of the antibody, modification of the antibody isotype to an isotype that naturally has reduced or eliminated effector function, and modification of the Fc region.
  • the effector function is reduced or eliminated by reduction of glycosylation of the antibody.
  • the glycosylation of the antibody is reduced by a method selected from: production of the antibody in an environment that does not permit wild-type glycosylation; removal of carbohydrate groups already present on the antibody; and modification of the antibody such that wild-type glycosylation does not occur.
  • the glycosylation of the antibody is reduced by a production of the antibody in an environment that does not permit wild-type glycosylation, such as production in a non- mammalian cell production system or where the antibody is produced synthetically.
  • the antibody is produced in a non-mammalian cell production system.
  • the antibody is produced synthetically.
  • the glycosylation of the antibody is reduced by modification of the antibody such that wild-type glycosylation does not occur, such as wherein the Fc region of the antibody comprises a mutation at position 297 such that the wild-type asparagine residue at that position is replaced with another amino acid that interferes with glycosylation at that position.
  • the effector function is reduced or eliminated by at least one modification of the Fc region.
  • the effector function or complement activation function is reduced or eliminated by deletion of all or a portion of the Fc region, or by engineering the antibody such that it does not include an Fc region or non-Fc region competent for effector function or complement activation function.
  • the at least one modification of the Fc region is selected from: a point mutation of the Fc region to impair binding to one or more Fc receptors selected from the following positions: 238, 239, 248, 249, 252, 254, 265, 268, 269, 270, 272, 278, 289, 292, 293, 294, 295, 296, 297, 298, 301 , 303, 322, 324, 327, 329, 333, 335, 338, 340, 373, 376, 382, 388, 389, 414, 416, 419, 434, 435, 437, 438, and 439; a point mutation of the Fc region to impair binding to Clq selected from the following positions: 270, 322, 329, and 321; eliminating some or all of the Fc region, and a point mutation at position 132 of the CHI domain.
  • the modification is a point mutation of the Fc region to impair binding to Clq selected from the following positions: 270, 322, 329, and 321.
  • the modification is elimination of some or all of the Fc region.
  • complement-triggering function is reduced or eliminated by deletion of all or a portion of the Fc region, or by engineering the antibody such that it does not include an Fc region that engages the complement pathway.
  • the antibody is selected from a Fab or a single chain antibody.
  • the non- Fc region of the antibody is modified to reduce or eliminate activation of the complement pathway by the antibody.
  • the modification is a point mutation of the CHI region to impair binding to C3.
  • the point mutation is at position 132 (see, e.g., Vidarte et al, (2001) J. Biol. Chem. 276(41): 38217-38223).
  • the antibody the half-life of the antibody is increased by a modification in the FcRn binding region.
  • the modification is a substitution in an amino acid selected from the following positions: 251 256, 285, 290, 308, 314, 385, 389, 428, 434, 436, 238, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434.
  • the modification is a substitution selected from the following: M252Y, S254T, T256E, N434A and Y436I.
  • the antibody is combined with a further compound that mitigates or contributes to the reduction of impact on reticulocyte levels or acute clinical symptoms.
  • the further compound protects reticulocytes from antibody-related depletion or supports the growth, development, or reestablishment of reticulocytes.
  • the further compound is selected from erythropoietin (EPO), an iron supplement, vitamin C, folic acid, and vitamin B 12, or is red blood cells or reticulocytes.
  • the affinity of the antibody for TfR is decreased, as measured relative to a wild-type antibody of the same isotype not having lowered affinity for TfR.
  • the antibody has a KD or IC50 for TfR of about 1 pM to about 100 ⁇ .
  • the dose amount and/or frequency of administration of the antibody is modulated to reduce the concentration of the antibody to which the red blood cells are exposed.
  • the antibody is coupled to a therapeutic compound. In another aspect of the above embodiment, the antibody is coupled to an imaging agent or a label. In one such aspect, the antibody is a multispecific antibody and the therapeutic compound optionally forms one portion of the multispecific antibody. In one such aspect, the multispecific antibody comprises a first antigen binding site which binds TfR and a second antigen binding site which binds a brain antigen.
  • the brain antigen is selected from the group consisting of: beta-secretase 1 (BACEl), Abeta, epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), tau, apolipoprotein E (ApoE), alpha-synuclein, CD20, huntingtin, prion protein (PrP), leucine rich repeat kinase 2 (LRRK2), parkin, presenilin 1, presenilin 2, gamma secretase, death receptor 6 (DR6), amyloid precursor protein (APP), p75 neurotropism receptor (p75NTR), and caspase 6.
  • the multispecific antibody binds both TfR and BACE1.
  • the multispecific antibody binds both TfR and Abeta.
  • the therapeutic compound is a neurological disorder drug.
  • the invention provides an isolated nucleic acid encoding any of the foregoing antibodies.
  • the invention provides a host cell comprising such nucleic acid.
  • the invention provides a method of producing any of the foregoing antibodies comprising culturing such host cell so that the antibody is produced and optionally further comprising recovering the antibody from the host cell.
  • the invention provides a pharmaceutical formulation comprising any of the foregoing antibodies and a pharmaceutically acceptable carrier.
  • the invention provides any of the foregoing antibodies for use as a medicament. In another aspect of the above embodiment, the invention provides the use of any of the foregoing antibodies in the manufacture of a medicament for treating a neurological disorder.
  • the neurological disorder is selected from the group consisting of a neuropathy disorder, a neurodegenerative disease, cancer, an ocular disease disorder, a seizure disorder, a lysosomal storage disease, amyloidosis, a viral or microbial disease, ischemia, a behavioral disorder, and CNS inflammation.
  • the invention provides any of the foregoing antibodies for use in treating a neurological disorder.
  • the neurological disorder is selected from the group consisting of a neuropathy disorder, a neurodegenerative disease, cancer, an ocular disease disorder, a seizure disorder, a lysosomal storage disease, amyloidosis, a viral or microbial disease, ischemia, a behavioral disorder, and CNS inflammation.
  • the invention provides any of the foregoing antibodies for use in transporting one or more compounds across the BBB.
  • use of any of the foregoing antibodies in the manufacture of a medicament for transporting one or more compounds across the BBB is provided.
  • a method of transporting a compound across the BBB in a subject comprising exposing any of the foregoing antibodies to the BBB such that the antibody transports the compound coupled thereto across the BBB.
  • the BBB is in a human subject.
  • the dose amount and/or frequency of administration is modulated to reduce the concentration of antibody to which red blood cells are exposed.
  • the method further comprises the step of monitoring the subject for depletion of red blood cells.
  • the antibody coupled to the compound is administered at a therapeutic dose.
  • the therapeutic dose is TfR-saturating.
  • administration of the antibody is at a dose and/or dose frequency calibrated to minimize acute clinical symptoms of the antibody administration.
  • a method of increasing exposure of the CNS of a subject to a compound comprising exposing any of the foregoing antibodies to the BBB such that the antibody transports the compound coupled thereto across the BBB.
  • the BBB is in a human subject.
  • the dose amount and/or frequency of administration is modulated to reduce the concentration of antibody to which red blood cells are exposed.
  • the method further comprises the step of monitoring the subject for depletion of red blood cells.
  • the antibody coupled to the compound is administered at a therapeutic dose.
  • the therapeutic dose is TfR-saturating.
  • administration of the antibody is at a dose and/or dose frequency calibrated to minimize acute clinical symptoms of the antibody administration.
  • a method of increasing rentention in the CNS of a compound administered to a subject comprising exposing any of the foregoing antibodies to the BBB such that the retention in the CNS of the compound is increased.
  • the BBB is in a human subject.
  • the dose amount and/or frequency of administration is modulated to reduce the concentration of antibody to which red blood cells are exposed.
  • the method further comprises the step of monitoring the subject for depletion of red blood cells.
  • the antibody coupled to the compound is administered at a therapeutic dose.
  • the therapeutic dose is TfR-saturating.
  • administration of the antibody is at a dose and/or dose frequency calibrated to minimize acute clinical symptoms of the antibody administration.
  • a method of treating a neurological disorder in a mammal comprising treating the mammal with any of the foregoing antibodies.
  • the neurological disorder is selected from the group consisting of a neuropathy disorder, a neurodegenerative disease, cancer, an ocular disease disorder, a seizure disorder, a lysosomal storage disease, amyloidosis, a viral or microbial disease, ischemia, a behavioral disorder, and CNS inflammation.
  • the neurological disorder is in a human subject.
  • the dose amount and/or frequency of administration is modulated to reduce the concentration of antibody to which the re blood cells are exposed.
  • the method further comprises the step of monitoring the subject for depletion of red blood cells.
  • the antibody coupled to the compound is administered at a therapeutic dose.
  • the therapeutic dose is TfR-saturating.
  • administration of the antibody is at a dose and/or dose frequency calibrated to minimize acute clinical symptoms of the antibody administration.
  • the invention provides an isolated antibody that binds to the same epitope on TfR as an antibody selected from the group consisting of antibodies 7A4, 8A2, 15D2, 10D11, 7B10, 15G11, 16G5, 13C3, 16G4, 16F6, 7G7, 4C2, 1B12, 13D4, 15Gl l .vl, 15Gl l .v2, 15G11.V3, 15Gl l .v4, 15Gl l.v5, 7A4.vl; 7A4.v2, 7A4.v3, 7A4.v4, 7A4.v5, 7A4.v6, 7A4.v7, 7A4.v8, 7A4.v9, 7A4.vlO, 7A4.vl l, 7A4.vl2, 7A4.vl3, 7A4.vl4, 7A4.vl5, 7G7.vl, 16F6.vl, 16F6.v2, 16F6.v3, 16F6.v4 15G11.N52A, 15G11.T53A
  • a method of decreasing clearance of a compound administered to a subject wherein the compound is coupled to an antibody which binds with low affinity to TfR, such that the clearance of the compound is decreased, and wherein reduction of red blood cell levels in the subject upon compound-coupled antibody
  • administration to the subject is decreased or eliminated.
  • pharmacodynamics of a compound to be efficacious in the CNS in a subject wherein the compound is coupled to an antibody which binds with low affinity to TfR, and the antibody is selected such that its affinity for TfR after coupling to the compound results in an amount of transport of the antibody conjugated to the compound across the BBB that optimizes the pharmacokinetics and/or pharmacodynamics of the compound in the CNS, wherein reduction of red blood cell levels in the subject upon compound-coupled antibody administration to the subject is decreased or eliminated.
  • Figure 1 depicts a three-dimensional crystal structure of a TfR dimer in complex with Tf, based on the pdb file 3SM9. The non-Tf-binding apical region of TfR is labeled.
  • Figures 2A-2B depict FACS analysis of mouse hybridoma parental clone supernatants binding to human and cynomolgus TfR transiently expressed in 293 cells in the presence of 1 ⁇ human holo-Tf.
  • the filled grey trace in each graph is background from the detection antibody
  • the medium grey trace is binding to 293 cells that endogenously express basal levels of human TfR
  • the bold black trace represents binding to transiently expressed human TfR
  • the thin grey trace represents binding to transiently expressed cyno TfR.
  • Figure 2C depicts the results of human/cynomolgous cross-reactive antibody competition assays as described in Example 1.
  • Nine of the fourteen clones were found to block binding of the apical binding antibody displayed on phage.
  • Figures 3A-1, 3A-2, 3B-1 , 3B-2, 3C-1 , 3C-2, 3D-land 3D-2 depict the heavy and light chain variable region sequences of hybridoma clones that bind to apical and non-apical regions of TfR.
  • the sequences can be further subdivided by epitope and sequence similarity into class I-III (apical binders) and class IV (non-apical binders).
  • the HVRs according to Kabat are indicated by underlining.
  • Each mouse light or heavy variable domain sequence (second line) is aligned to the closest human germline or consensus variable domain (first line).
  • the humanized version for each antibody is shown at the bottom (third line). Differences from the human germline or consensus sequences are shaded.
  • HVR sequences that were grafted into the human framework are boxed. CDR definitions according to Kabat are indicated.
  • Figure 4E-1 and 4E-2 show that for the class I-III groups of antibodies, variant forms of the antibodies with modifications at one or more residues of an FR retained affinity and binding specifity.
  • Figure 5 depicts the binding of hu7A4.vl5, hul5Gl l .v5 and hu7G7.vl to huTfR in the presence of 6.3 ⁇ holo-Tf.
  • Antibody binding to immobilized huTfR is shown in the presence (open sybols and dashed lines) or absence (filled symbols and solid lines) of 6.3 ⁇ holo-Tf.
  • Figures 6A-B depict the results of the HFE - HuTfR binding and the HFE blocking assays described in Example 1.
  • Figure 6A shows the binding of antibody to increasing concentrations of huTfR captured via immobilized HFE.
  • Figure 6B shows the binding of huTfR to immobilized HFE in the presence of increasing concentrations of antibody.
  • Figure 7A-B depict binding analyses of 15G11.v5 and 7A4.v5 IgG and Fab Ala variants on cyno and human TfR, demonstrating the effects on affinity of Ala mutations in CDR-L3 and CDR-H3 of each antibody assessed as IgG by ELISA binding and IgG or Fab by SPR analysis to immobilized human or cyno TfR, as described in Example 2.
  • FIGs 8A-B and Figures 9A-B depict the results of experiments assessing the impact of effector function status on ADCC activity of anti-human TfR ("anti-hTFR”) antibodies in primary human bone marrow mononuclear cells or in a human erythroblast cell line, as described in Example 4.
  • anti-hTFR anti-human TfR
  • Figure 10 depicts the dosing and sampling scheme for the primate study described in Example .
  • Figures 11A-11B depict the pharmacokinetic results of the experiments described in Example 5, specifically individual and group mean anti-TfRVBACEl, anti-TfR 2 /BACE 1 and anti-gD serum concentrations versus time following a single TV bolus administration at 30 mg/kg in cynomolgus monkeys in serum ( Figure 11 A) and CSF ( Figure 1 IB).
  • Figures 12A-12E depict the pharmacodynamic results of the experiments described in
  • Example 5 specifically individual and group mean anti-TfRVBACEl, anti-TfR 2 /BACE 1 and anti-gD plasma (A) or CSF (B-E) concentrations versus time following a single IV bolus administration at 30 mg kg in cynomolgus monkeys.
  • the upper panels show Abetal-40 levels in plasma (Figure 12A) and CSF (Figure 12B), while the lower panels show soluble APP levels ( Figure 12C), soluble ⁇ levels ( Figure 12D), and sAPPp/sAPPa ratio (Figure 12E) over time.
  • Figures 13A-13D depict the results of hematological sampling performed during the studies described in Example 5. At each of the indicated time points, total reticulocytes (Figure 13 A), red blood cells (Figure 13B), hemoglobin (Figure 13D) and the percentage of immature reticulocytes in the total reticulocyte pool ( Figure 13C) were measured using standard techniques.
  • Figure 14 depicts the dosing and sampling scheme for the primate study described in Example 6.
  • Figures 15A-15B depict the pharmacodynamic results (A) and brain antibody concentrations (B) of the experiments described in Example 6.
  • Figure 15A shows individual and group mean anti-TfR /BACEl, anti-TfRVBACEl , anti-gD, and anti- BACE1 ration of sAPPp/sAPPa in CSF versus time following a single IV bolus administration at 30 mg/kg in cynomolgus monkeys.
  • Figure 15B show individual. anti-TfRVBACEl, anti- Tffi ' /BACEl, anti-gD, and anti-BACEl concentrations of antibody in various brain regions at 24 hours post-dose.
  • Figures 16A-B depict the light and heavy chain amino acid sequences of anti-B ACE 1 clone YW412.8 obtained from a na ' ive sort of the natural diversity phage display library and affinity-matured forms of YW412.8.
  • Fig. 16A depicts the variable light (VL) sequence alignments (SEQ ID NOs. 132-137).
  • Fig. 16B depicts the variable heavy (VH) sequence alignments (SEQ ID Nos. 138-139).
  • the HVR sequences for each clone are indicated by the boxed regions, with the first box indicating HVR-L1 (Fig. 16A) or HVR-H1 (Fig. 16B), the second box indicating HVR-L2 (Fig. 16A) or HVR-H2 (Fig. 16B), and the third box indicating HVR-L3 (Fig. 16A) or HVR-H3 (Fig. 16B).
  • Figures 17A-B depict the light and heavy chain amino acid sequences of anti-B ACE 1 antibody clone Fab 12 obtained from a na ' ive sort of a synthetic diversity phage display library and affinity-matured forms of Fab 12.
  • Fig. 17A depicts the light chain sequence alignments (SEQ ID NOs. 140-143).
  • Fig. 17B depicts the heavy chain sequence alignments (SEQ ID NO. 144).
  • the HVR sequences for each clone are indicated by the boxed regions, with the first box indicating HVR-L1 (Fig. 17A) or HVR-Hl (Fig. 17B), the second box indicating HVR-L2 (Fig. 17A) or HVR-H2 (Fig. 17B), and the third box indicating HVR-L3 (Fig. 17A) or HVR-H3 (Fig. 17B).
  • Figures 18A-B depict the heavy chain (Fig. 18A; SEQ ID NO. 145) and light chain (Fig. 18B; SEQ ID NO. 146) of an exemplary anti-Abeta antibody.
  • Figure 19 depicts the pharmacokinetic properties of Anti-TfRVBACEl, Anti- Tfr 52A /BACEl and Anti-TfR 53A /BACE 1 as described in Example 5.
  • Figure 20 depicts the pharmacokinetic properties of the murine IgG2a Anti- TfR D /BACEl and Anti-gD antibodies with the Fc effector function LALAPG mutations as described in Example 7.
  • Figure 21 depicts the total and immature reticulocyte count in mice 24 hours after administration of a 50mg/kg dose of the murine IgG2a Anti-TfR D /BACEl and Anti-gD antibodies with the Fc effector function LALAPG mutations as described in Example 7.
  • Figure 22 depicts the total reticulocyte count in mice 24 hours after administration of a 50mg/kg dose of the anti-TfR 52A /BACEl (N297G), anti-TfR 52A /BACE 1 (LALAPG), anti- TfR 52 A/BACEl (LALAPG/YTE), TfR 52 A/BACEl (LALAPG/AI) antibodies in human transferrin receptor knock-in mice as described in Example 8.
  • Figure 23 depicts the results of experiments assessing the impact of effector function status on ADCC activity of anti-TfR/gD, anti-TfR/BACEl (N297G), anti-TfR/BACEl (LALAPG), anti-TfR/BACE 1 (N297G/434A/436I) and anti-TfR/BACE 1 (LALAPG/YTE) antibodies in primary human bone marrow mononuclear cells or in a human erythroblast cell line, as described in Example 8.
  • Figure 24 depicts the heavy and light chain variable region sequences of 151 lGv.5 (light chain - SEQ ID NO: 105 and heavy chain - SEQ ID NO: 108) and affinity variants
  • 15G11.52A (light chain - SEQ ID NO: 105 and heavy chain - SEQ ID NO: 153), 15G11.53A (light chain - SEQ ID NO: 105 and heavy chain - SEQ ID NO: 154) and 15G11.92A (light chain - SEQ ID NO: 151 and heavy chain - SEQ ID NO: 108).
  • the HVRs according to Kabat are indicated by underlining.
  • Figure 25 depicts a competition assay between 15G1 lv.5 and anti-TfR as described in Example 1.
  • Binding affinity refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD, which is a ratio of the off-rate of X from Y (kd or Koff) to the on-rate of X to Y (ka or kon)).
  • a surrogate measurement for the affinity of one or more antibodies for its target is its half maximal inhibitory concentration (IC50), a measure of how much of the antibody is needed to inhibit the binding of a known ligand to the antibody target by 50%.
  • IC50 half maximal inhibitory concentration
  • Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described herein.
  • the "blood-brain barrier” or “BBB” refers to the physiological barrier between the peripheral circulation and the brain and spinal cord (i.e., the CNS) which is formed by tight junctions within the brain capillary endothelial plasma membranes, creating a tight barrier that restricts the transport of molecules into the brain, even very small molecules such as urea (60 Daltons).
  • the blood-brain barrier within the brain, the blood-spinal cord barrier within the spinal cord, and the blood-retinal barrier within the retina are contiguous capillary barriers within the CNS, and are herein collectively referred to a the blood-brain barrier or BBB.
  • the BBB also encompasses the blood-CSF barrier (choroid plexus) where the barrier is comprised of ependymal cells rather than capillary endothelial cells.
  • amyloid beta refers to the fragment of amyloid precursor protein (“APP) that is produced upon ⁇ -secretase 1 (“BACEl”) cleavage of APP, as well as modifications, fragments and any functional equivalents thereof, including, but not limited to, ⁇ _4 ⁇ , and ⁇ _4 2 .
  • APP amyloid precursor protein
  • BACEl ⁇ -secretase 1
  • is known to exist in monomeric form, as well as to associate to form oligomers and fibril structures, which may be found as constituent members of amyloid plaque.
  • ⁇ peptides are well known to one of ordinary skill in the art and methods of producing said peptides or of extracting them from brain and other tissues are described, for example, in Glenner and Wong, Biochem Biophys Res. Comm. 129: 885-890 (1984).
  • ⁇ peptides are also commercially available in various forms.
  • Anti-Abeta immunoglobulin refers to an antibody that specifically binds to human Abeta.
  • a nonlimiting example of an anti-Abeta antibody is crenezumab.
  • Other non-limiting examples of anti-Abeta antibodies are solanezumab, bapineuzumab, gantenerumab, aducanumab, ponezumab
  • crenezumab and “MABT5102 A” are used interchangeably herein, and refer to a specific anti-Abeta antibody that binds to monomeric, oligomeric, and fibril forms of Abeta, and which is associated with CAS registry number 1095207.
  • such antibody comprises sequences set forth in Figures 18A and 18B.
  • Apolipoprotein E4 carrier or “ApoE4 carrier,” used interchangeably herein with
  • apolipoprotein E4 positive or "ApoE4 positive,” refers to an individual having at least one apolipoprotein E4 (or "ApoE4") allele.
  • An individual with zero ApoE4 alleles is referred to herein as being "ApoE4 negative” or an "ApoE4 non-carrier.” See also Prekumar, et al., 1996, Am. J Pathol. 148:2083-95.
  • Cerebral vasogenic edema refers to an excess accumulation of intravascular fluid or protein in the intracellular or extracellular spaces of the brain. Cerebral vasogenic edema is detectable by, e.g., brain MRI, including, but not limited to FLAIR MRI, and can be asymptomatic ("asymptomatic vasogenic edema") or associated with neurological symptoms, such as confusion, dizziness, vomiting, and lethargy (“symptomatic vasogenic edema”) (see Sperling et al. Alzheimer 's & Dementia, 7:367, 2011).
  • Cerebral macrohemorrhage refers to an intracranial hemorrhage, or bleeding in the brain, of an area that is more than about 1 cm in diameter. Cerebral macrohemorrhage is detectable by, e.g., brain MRI, including but not limited to T2* -weighted GRE MRI, and can be asymptomatic ("asymptomatic macrohemorrhage") or associated with symptoms such as transient or permanent focal motor or sensory impairment, ataxia, aphasia, and dysarthria (“symptomatic macrohemorrhage”) (see, e.g., Chalela JA, Gomes J. Expert Rev. Neurother. 2004 4:267, 2004 and Sperling et al. Alzheimer's & Dementia, 7:367, 2011).
  • asymptomatic macrohemorrhage e.g., Chalela JA, Gomes J. Expert Rev. Neurother. 2004 4:267, 2004 and Sperling et al.
  • Cerebral microhemorrhage refers to an intracranial hemorrhage, or bleeding in the brain, of an area that is less than about 1 cm in diameter. Cerebral microhemorrhage is detectable by, e.g., brain MRI, including, but not limited to T2* -weighted GRE MRI, and can be asymptomatic ("asymptomatic microhemorrhage") or can potentially be associated with symptoms such as transient or permanent focal motor or sensory impairment, ataxia, aphasia, and dysarthria ("symptomatic microhemorrhage"). See, e.g., Greenberg, et al., 2009, Lancet Neurol. 8: 165-74.
  • sulcal effusion refers to an effusion of fluid in the furrows, or sulci, of the brain. Sulcal effusions are detectable by, e.g., brain MRI, including but not limited to FLAIR MRI. Seesperling et al. Alzheimer 's & Dementia, 7:367, 2011.
  • the term "superficial siderosis of the central nervous system” refers to bleeding or hemorrhage into the subarachnoid space of the brain and is detectable by, e.g., brain MRI, including but not limited to T2* -weighted GRE MRI. Symptoms indicative of superficial siderosis of the central nervous system include sensorineural deafness, cerebellar ataxia, and pyramidal signs. See Kumara-N, Am J Neuroradiol. 31 :5, 2010.
  • amyloidosis refers to a group of diseases and disorders caused by or associated with amyloid or amyloid-like proteins and includes, but is not limited to, diseases and disorders caused by the presence or activity of amyloid-like proteins in monomeric, fibril, or polymeric state, or any combination of the three, including by amyloid plaques.
  • diseases include, but are not limited to, secondary amyloidosis and age-related amyloidosis, such as diseases including, but not limited to, neurological disorders such as Alzheimer's
  • AD Alzheimer's disease
  • MCI mild cognitive impairment
  • Lewy body dementia Lewy body dementia
  • Down's syndrome hereditary cerebral hemorrhage with amyloidosis
  • Dutch type hereditary cerebral hemorrhage with amyloidosis
  • Parkinson-Demential complex and other diseases which are based on or associated with amyloid-like proteins such as progressive supranuclear palsy, multiple sclerosis, Creutzfeld Jacob disease, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), inclusion-body myositis (IBM), adult onset diabetes, endocrine tumor and senile cardiac amyloidosis, and various eye diseases including macular degeneration, drusen-related optic neuropathy, glaucoma, and cataract due to beta-amyloid deposition.
  • amyloid-like proteins such as progressive supranuclear palsy, multiple sclerosis, Creutzfeld Jacob disease, Parkinson's disease, HIV-related dementia, ALS (amyotropic lateral sclerosis), inclusion-body myositis (IBM), adult onset diabetes, endocrine tumor and senile cardiac amyloidosis, and various eye diseases including macular degeneration, drusen-related optic neuropathy, glau
  • Glaucoma is a group of diseases of the optic nerve involving loss of retinal ganglion cells (RGCs) in a characteristic pattern of optic neuropathy.
  • RGCs are the nerve cells that transmit visual signals from the eye to the brain.
  • Caspase-3 and Caspase-8 two major enzymes in the apoptotic process, are activated in the process leading to apoptosis of RGCs.
  • Caspase-3 cleaves amyloid precursor protein (APP) to produce neurotoxic fragments, including Abeta. Without the protective effect of APP, Abeta accumulation in the retinal ganglion cell layer results in the death of RGCs and irreversible loss of vision.
  • APP amyloid precursor protein
  • Glaucoma is often, but not always, accompanied by an increased eye pressure, which may be a result of blockage of the circulation of aqueous, or its drainage.
  • raised intraocular pressure is a significant risk factor for developing glaucoma
  • no threshold of intraocular pressure can be defined which would be determinative for causing glaucoma.
  • the damage may also be caused by poor blood supply to the vital optic nerve fibers, a weakness in the structure of the nerve, and/or a problem in the health of the nerve fibers themselves.
  • Untreated glaucoma leads to permanent damage of the optic nerve and resultant visual field loss, which can progress to blindness.
  • micro Alzheimer's Disease or "mild AD” as used herein (e.g., a “patient diagnosed with mild AD”) refers to a stage of AD characterized by an MMSE score of 20 to 26.
  • Mild to moderate Alzheimer's Disease or “mild to moderate AD” as used herein encompasses both mild and moderate AD, and is characterized by an MMSE score of 18 to 26.
  • moderate Alzheimer's Disease or “moderate AD” as used herein (e.g. , a)
  • patient diagnosed with moderate AD refers to a stage of AD characterized by an MMSE score of 18 to 19.
  • central nervous system or “CNS” refers to the complex of nerve tissues that control bodily function, and includes the brain and spinal cord.
  • BBB-R blood-brain barrier receptor
  • IGF-R insulin-like growth factor receptor
  • LRP8 low density lipoprotein receptors including without limitation low density lipoprotein receptor-related protein 1 (LR l) and low density lipoprotein receptor-related protein 8 (LRP8), glucose transporter 1 (Glutl) and heparin-binding epidermal growth factor-like growth factor (HB- EGF).
  • TfR transferrin receptor
  • IGF-R insulin-like growth factor receptor
  • LRP8 low density lipoprotein receptors
  • Glutl glucose transporter 1
  • HB- EGF heparin-binding epidermal growth factor-like growth factor
  • TfR transferrin receptor
  • mammals such as primates (e.g., humans) and rodents (e.g., mice and rats), unless otherwise indicated.
  • the term encompasses "full-length,” unprocessed TfR as well as any form of TfR that results from processing in the cell.
  • the term also encompasses naturally occurring variants of TfR, e.g., splice variants or allelic variants.
  • TfR is a transmembrane glycoprotein (with a molecular weight of about 180,000) composed of two disulphide-bonded sub-units (each of apparent molecular weight of about 90,000) involved in iron uptake in vertebrates.
  • the TfR herein is human TfR ("hTfR") comprising the amino acid sequence as set forth in Schneider et al. Nature 311 : 675 - 678 (1984), for example (SEQ ID NO: 1).
  • the TfR herein is primate TfR ("pTfR") comprising the amino acid sequence as set forth in Genbank reference AFD 18260.1 (SEQ ID NO: 2).
  • pTfR primate TfR
  • the mouse TfR sequence may be found in Genbank reference AAH54522.1 (SEQ ID NO: 3).
  • a “neurological disorder” as used herein refers to a disease or disorder which affects the CNS and/or which has an etiology in the CNS.
  • exemplary CNS diseases or disorders include, but are not limited to, neuropathy, amyloidosis, cancer, an ocular disease or disorder, viral or microbial infection, inflammation, ischemia, neurodegenerative disease, seizure, behavioral disorders, and a lysosomal storage disease.
  • the CNS will be understood to include the eye, which is normally sequestered from the rest of the body by the blood-retina barrier.
  • neurological disorders include, but are not limited to, neurodegenerative diseases (including, but not limited to, Lewy body disease, postpoliomyelitis syndrome, Shy-Draeger syndrome, olivopontocerebellar atrophy, Parkinson's disease, multiple system atrophy, striatonigral degeneration, tauopathies (including, but not limited to, Alzheimer disease and supranuclear palsy), prion diseases (including, but not limited to, bovine spongiform encephalopathy, scrapie, Creutzfeldt- Jakob syndrome, kuru, Gerstmann-Straussler-Scheinker disease, chronic wasting disease, and fatal familial insomnia), bulbar palsy, motor neuron disease, and nervous system heterodegenerative disorders
  • neurodegenerative diseases including, but not limited to, Lewy body disease, postpoliomyelitis syndrome, Shy-Draeger syndrome, olivopontocerebellar atrophy, Parkinson's disease, multiple system atrophy, striatonigral degeneration, tauopathies
  • hepatolenticular degeneration e.g., Lesch-Nyhan syndrome, and Unverricht-Lundborg syndrome
  • dementia including, but not limited to, Pick's disease, and spinocerebellar ataxia
  • cancer e.g. of the CNS, including brain metastases resulting from cancer elsewhere in the body.
  • Neurological disorder drug is a drug or therapeutic agent that treats one or more neurological disorder(s).
  • Neurological disorder drugs of the invention include, but are not limited to, antibodies, peptides, proteins, natural ligands of one or more CNS target(s), modified versions of natural ligands of one or more CNS target(s), aptamers, inhibitory nucleic acids (i.e., small inhibitory RNAs (siRNA) and short hairpin RNAs (shRNA)), ribozymes, and small molecules, or active fragments of any of the foregoing.
  • siRNA small inhibitory RNAs
  • shRNA short hairpin RNAs
  • Exemplary neurological disorder drugs of the invention include, but are not limited to: antibodies, aptamers, proteins, peptides, inhibitory nucleic acids and small molecules and active fragments of any of the foregoing that either are themselves or specifically recognize and/or act upon (i.e., inhibit, activate, or detect) a CNS antigen or target molecule such as, but not limited to, amyloid precursor protein or portions thereof, amyloid beta, beta-secretase, gamma-secretase, tau, alpha-synuclein, parkin, huntingtin, DR6, presenilin, ApoE, glioma or other CNS cancer markers, and neurotrophins.
  • Non-limiting examples of neurological disorder drugs and the disorders they may be used to treat are provided in the following Table 1 : TABLE 1 :
  • Non-limiting examples of neurological disorder drugs and the corresponding disorders they may be used to treat are provided in the following Table 1 : TABLE 1 :
  • BDNF Brain-derived neurotrophic factor
  • CNTF Ciliary neurotrophic factor
  • Anti-HER2 antibody e.g. trastuzumab, Brain metastasis from HER2 -positive pertuzumab, etc.
  • cancer e.g. trastuzumab, Brain metastasis from HER2 -positive pertuzumab, etc.
  • Anti-VEGF antibody e.g., bevacizumab Recurrent or newly diagnosed
  • glioblastoma recurrent malignant glioma
  • brain metastasis glioblastoma, recurrent malignant glioma, brain metastasis
  • imaging agent is a compound that has one or more properties that permit its presence and/or location to be detected directly or indirectly.
  • imaging agents include proteins and small molecule compounds incorporating a labeled moiety that permits detection.
  • a “CNS antigen” or “brain antigen” is an antigen expressed in the CNS, including the brain, which can be targeted with an antibody or small molecule.
  • antigens include, without limitation: beta-secretase 1 (BACEl), amyloid beta (Abeta), epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), tau, apolipoprotein E4 (ApoE4), alpha-synuclein, CD20, huntingtin, prion protein (PrP), leucine rich repeat kinase 2 (LRRK2), parkin, presenilin 1 , presenilin 2, gamma secretase, death receptor 6 (DR6), amyloid precursor protein (APP), p75 neurotrophin receptor (p75NTR), interleukin 6 receptor (IL6R), TNF receptor 1 (TNFR1), interleukin 1 beta (IL1 ⁇ ), and caspase 6.
  • BACEl beta-secretase 1
  • the antigen is BACEl .
  • BACEl refers to any native beta-secretase 1 (also called ⁇ - site amyloid precursor protein cleaving enzyme I , membrane-associated aspartic protease 2, memapsin 2, aspartyl protease 2 or Asp2) from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g., mice and rats), unless otherwise indicated.
  • the term encompasses "full-length,” unprocessed BACEl as well as any form of BACEl which results from processing in the cell.
  • the term also encompasses naturally occurring variants of BACE 1 , e.g. , splice variants or allelic variants.
  • the amino acid sequence of an exemplary BACEl polypeptide is the sequence for human BACEl, isoform A as reported in Vassar et al, Science 286:735-741 (1999), which is incorporated herein by reference in its entirety.
  • isoforms B, C and D See UniProtKB/Swiss- Prot Entry P56817, which is incorporated herein by reference in its entirety.
  • anti-beta-secretase antibody refers to an antibody that is capable of binding BACEl with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting BACEl .
  • the extent of binding of an anti-BACEl antibody to an unrelated, non-BACEl protein is less than about 10% of the binding of the antibody to BACEl as measured, e.g., by a radioimmunoassay (RIA).
  • an antibody that binds to BACEl has a dissociation constant (Kd) of ⁇ ⁇ ⁇ , ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g. 10 8 M or less, e.g. from 10 ⁇ 8 M to 10 "13 M, e.g., from 10 "9 M to 10 "13 M).
  • Kd dissociation constant
  • an anti-BACEl antibody binds to an epitope of BACEl that is conserved among BACEl from different species and isoforms.
  • an antibody that binds to the epitope on BACEl bound by anti-BACE 1 antibody YW412.8.31. In other embodiments, an antibody is provided that binds to an exosite within BACEl located in the catalytic domain of BACEl . In one embodiment an antibody is provided that competes with the peptides identified in Kornacker et al., Biochem.
  • Exemplary BACEl antibody sequences are depicted in Fig. 15A-B and Fig. 16A-B.
  • One exemplary antibody herein comprises the variable domains of the antibody YW412.8.31 (e.g. as in Figs. 15A-B).
  • a “native sequence” protein herein refers to a protein comprising the amino acid sequence of a protein found in nature, including naturally occurring variants of the protein.
  • the term as used herein includes the protein as isolated from a natural source thereof or as recombinantly produced.
  • antibody herein is used in the broadest sense and encompasses various antibody stuctures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
  • antibody fragment refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments are well known in the art (see, e.g., Nelson, MAbs (2010) 2(1): 77-83) and include but are not limited to Fab, Fab', Fab'-SH, F(ab') 2 , and Fv; diabodies; linear antibodies; single-chain antibody molecules including but not limited to single-chain variable fragments (scFv), fusions of light and/or heavy-chain antigen-binding domains with or without a linker (and optionally in tandem); and monospecific or multispecific antigen-binding molecules formed from antibody fragments (including, but not limited to multispecific antibodies constructed from multiple variable domains which lack Fc regions).
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variants, e.g., containing naturally occurring mutations or that may arise during production of the monoclonal antibody, such variants generally being present in minor amounts.
  • polyclonal antibody preparations typically include different antibodies directed against different determinants (epitopes)
  • each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on the antigen.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridoma method (see, e.g., Kohler et al, Nature, 256:495 (1975)), recombinant DNA methods (see, e.g., U.S. Patent No.
  • phage-display methods e.g., using the techniques described in Clackson et al, Nature, 352:624-628 (1991) and Marks et al, J. Mol Biol, 222:581-597 (1991)
  • methods utilizing transgenic animals containing all or part of the human immunoglobulin loci such methods and other exemplary methods for making monoclonal antibodies being described herein.
  • monoclonal antibodies herein include chimeric antibodies, humanized antibodies, and human antibodies, including antigen-binding fragments thereof.
  • the monoclonal antibodies herein specifically include “chimeric” antibodies
  • immunoglobulins in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Patent No. 4,816,567; Morrison et al, Proc. Natl Acad. Sci. USA, 81 :6851-6855 (1984)).
  • acceptor human framework for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below.
  • An acceptor human framework "derived from” a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less.
  • the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
  • a "human consensus framework” is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences.
  • the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences.
  • the subgroup of sequences is a subgroup as in Kabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, NIH
  • the subgroup is subgroup kappa I as in Kabat et al., supra. In one embodiment, for the VH, the subgroup is subgroup III as in Kabat et al., supra.
  • Humanized forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human antibodies.
  • humanized antibodies are human antibodies (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g., CDRs) correspond to those of a non-human antibody, and all or substantially all of the framework regions (FRs) correspond to those of a human antibody.
  • HVRs e.g., CDRs
  • FRs framework regions
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable regions correspond to those of a non-human antibody and all or substantially all of the FRs are those of a human antibody, except for FR substitution(s) as noted above.
  • the humanized antibody optionally also will comprise at least a portion of an antibody constant region, typically that of a human antibody.
  • a "humanized form" of an antibody, e.g., a non-human antibody refers to an antibody that has undergone humanization. For further details, see Jones et al, Nature 321 :522-525 (1986); Riechmann et al, Nature 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol 2:593-596 (1992).
  • a "human antibody” herein is an antibody comprising an amino acid sequence structure that corresponds with the amino acid sequence structure of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
  • Such antibodies can be identified or made by a variety of techniques, including, but not limited to: production by transgenic animals (e.g., mice) that are capable, upon immunization, of producing human antibodies in the absence of endogenous immunoglobulin production (see, e.g., Jakobovits et al, Proc. Natl. Acad. Sci.
  • a “multispecific antibody” herein is an antibody having binding specificities for at least two different epitopes.
  • Exemplary multispecific antibodies may bind both a TfR and a brain antigen.
  • Multispecific antibodies can be prepared as full-length antibodies or antibody fragments ⁇ e.g. F(ab') 2 bispecific antibodies).
  • Engineered antibodies with two, three or more (e.g. four) functional antigen binding sites are also contemplated (see, e.g., US Appln No. US 2002/0004587 Al , Miller et al.).
  • Multispecific antibodies can be prepared as full length antibodies or as antibody fragments.
  • Antibodies herein include "amino acid sequence variants" with altered antigen-binding or biological activity.
  • amino acid alterations include antibodies with enhanced affinity for antigen (e.g. "affinity matured” antibodies), and antibodies with altered Fc region, if present, e.g. with altered (increased or diminished) antibody dependent cellular cytotoxicity (ADCC) and/or complement dependent cytotoxicity (CDC) (see, for example, WO 00/42072, Presta, L. and WO 99/51642, Iduosogie et al.); and/or increased or diminished serum half-life (see, for example, WOOO/42072, Presta, L.).
  • ADCC antibody dependent cellular cytotoxicity
  • CDC complement dependent cytotoxicity
  • an "affinity modified variant” has one or more substituted hypervariable region or framework residues of a parent antibody (e.g. of a parent chimeric, humanized, or human antibody) that alter (increase or reduce) affinity.
  • a parent antibody e.g. of a parent chimeric, humanized, or human antibody
  • a convenient way for generating such substitutional variants uses phage display. Briefly, several hypervariable region sites (e.g. 6-7 sites) are mutated to generate all possible amino substitutions at each site. The antibody variants thus generated are displayed in a monovalent fashion from filamentous phage particles as fusions to the gene III product of M13 packaged within each particle. The phage-displayed variants are then screened for their biological activity (e.g. binding affinity).
  • alanine scanning mutagenesis can be performed to identify hypervariable region residues contributing significantly to antigen binding.
  • a "pH-sensitive antibody variant” is an antibody variant which has a different binding binding affinity for a target antigen at a first pH than it does for that target antigen at a different pH.
  • an anti-TfR antibody of the invention may be selected for or engineered to have pH-sensitive binding to TfR such that it binds with desirably low affinity (as described herein) to cell surface TfR in the plasma at pH 7.4, but upon internalization into an endosomal compartment, rapidly dissociates from TfR at the relatively lower pH (pH 5.5- 6.0); such dissociation may protect the antibody from antigen-mediated clearance, and increase the amount of antibody that is either delivered to the CNS or recycled back across the BBB - in either case, the effective concentration of the antibody is increased relative to an anti-TfR antibody that does not comprise such pH sensitivity (see, e.g., Chaparro-Riggers et al.
  • the antibody herein may be conjugated with a "heterologous molecule" for example to increase half-life or stability or otherwise improve the antibody.
  • the antibody may be linked to one of a variety of non-proteinaceous polymers, e.g. , polyethylene glycol (PEG), polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol.
  • Antibody fragments, such as Fab', linked to one or more PEG molecules are an exemplary embodiment of the invention.
  • the heterologous molecule is a therapeutic compound or a visualization agent (ie., a detectable label), and the antibody is being used to transport such heterologous molecule across the BBB.
  • heterologous molecules include, but are not limited to, a chemical compound, a peptide, a polymer, a lipid, a nucleic acid, and a protein.
  • the antibody herein may be a "glycosylation variant" such that any carbohydrate attached to the Fc region, if present, is altered, either modified in presence/absence, or modified in type.
  • glycosylation variant such that any carbohydrate attached to the Fc region, if present, is altered, either modified in presence/absence, or modified in type.
  • antibodies with a mature carbohydrate structure that lacks fucose attached to an Fc region of the antibody are described in US Pat Appl No US
  • oligosaccharide attached to an Fc region of the antibody are reported in WO 1997/30087, Patel et al. See, also, WO 1998/58964 (Raju, S.) and WO 1999/22764 (Raju, S.) concerning antibodies with altered carbohydrate attached to the Fc region thereof. See also US 2014/30087, Patel et al. See, also, WO 1998/58964 (Raju, S.) and WO 1999/22764 (Raju, S.) concerning antibodies with altered carbohydrate attached to the Fc region thereof. See also US
  • hypervariable region refers to each of the regions of an antibody variable domain which are hypervariable in sequence ("complementarity determining regions” or “CDRs") and/or form structurally defined loops ("hypervariable loops") and/or contain the antigen-contacting residues ("antigen contact”).
  • CDRs complementarity determining regions
  • hypervariable loops form structurally defined loops
  • antigen contact Generally, antibodies comprise six HVRs: three in the VH (HI, H2, H3), and three in the VL (LI, L2, L3).
  • Exemplary HVRs herein include: (a) hypervariable loops occurring at amino acid residues 26-32 (LI), 50-52 (L2), 91-96 (L3), 26-32 (HI), 53-55 (H2), and 96-101 (H3) (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987));
  • HVR residues comprise those identified in Figures 3A-D or 4A-D, Table 4 or Table 5 or elsewhere in the specification.
  • HVR residues and other residues in the variable domain are Unless otherwise indicated, HVR residues and other residues in the variable domain
  • FR residues are those variable domain residues other than the hypervariable region residues as herein defined.
  • the FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3 and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1-H1(L1)-FR2-
  • one or more FR residue may be modified to modulate the stability of the antibody or to modulate the three-dimensional positioning of one or more HVR of the antibody to, e.g., enhance binding.
  • a “full length antibody” is one which comprises an antigen-binding variable region as well as a light chain constant domain (CL) and heavy chain constant domains, CHI, CH2 and CH3.
  • the constant domains may be native sequence constant domains (e.g. human native sequence constant domains) or amino acid sequence variants thereof.
  • full length antibody “intact antibody,” and “whole antibody” are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
  • a “naked antibody” refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety or radiolabel).
  • the naked antibody may be present in a pharmaceutical formulation.
  • “Native antibodies” refer to naturally occurring immunoglobulin molecules with varying structures. For example, native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CHI, CH2 and CH3).
  • VH variable region
  • each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain.
  • VL variable region
  • CL constant light
  • the light chain of an antibody may be assigned to one of two types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequence of its constant domain.
  • Antibody effector functions refer to those biological activities of an antibody that result in activation of the immune system other than activation of the complement pathway. Such activities are largely found in the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody. Examples of antibody effector functions include, for example, Fc receptor binding and antibody-dependent cell-mediated cytotoxicity (ADCC).
  • the antibody herein essentially lacks effector function. In another embodiment, the antibody herein retains minimal effector function.
  • Methods of modifying or eliminating effector function are well-known in the art and include, but are not limited to, eliminating all or a portion of the Fc region responsible for the effector function (ie, using an antibody or antibody fragment in a format lacking all or a portion of the Fc region such as, but not limited to, a Fab fragment, a single-chain antibody, and the like as described herein and as known in the art; modifying the Fc region at one or more amino acid positions to eliminate effector function (Fc binding-impacting: positions 238, 239, 248, 249, 252, 254, 256, 265, 268, 269, 270, 272, 278, 289, 292, 293, 294, 295, 296, 297, 298, 301, 303, 311, 322, 324, 327, 329, 333, 335, 338, 340, 373, 376, 382, 388, 389
  • Antibody “complement activation” functions, or properties of an antibody that enable or trigger “activation of the complement pathway” are used interchangeably, and refer to those biological activities of an antibody that engage or stimulate the complement pathway of the immune system in a subject. Such activities include, e.g., Clq binding and complement dependent cytotoxicity (CDC), and may be mediated by both the Fc portion and the non-Fc portion of the antibody.
  • CDC complement dependent cytotoxicity
  • Methods of modifying or eliminating complement activation function include, but are not limited to, eliminating all or a portion of the Fc region responsible for complement activation (ie., using an antibody or antibody fragment in a format lacking all or a portion of the Fc region such as, but not limited to, a Fab fragment, a single-chain antibody, and the like as described herein and as known in the art, or modifying the Fc region at one or more amino acid positions to eliminate or lessen interactions with complement components or the ability to activate complement components, such as positions 270, 322, 329 and 321, known to be involved in Clq binding), and modifying or eliminating a portion of the non-Fc region responsible for complement activation (ie, modifying the CHI region at position 132 (see, e.g., Vidarte et al., (2001) J. Biol. Chem. 276(41): 38217-38223)).
  • full length antibodies can be assigned to different "classes". There are five major classes of full length antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into “subclasses” (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgA, and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of antibodies are called alpha, delta, epsilon, gamma, and mu, respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known in the art.
  • recombinant antibody refers to an antibody (e.g. a chimeric, humanized, or human antibody or antigen-binding fragment thereof) that is expressed by a recombinant host cell comprising nucleic acid encoding the antibody.
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cells and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
  • Examples of "host cells” for producing recombinant antibodies include: (1) mammalian cells, for example, Chinese Hamster Ovary (CHO), COS, myeloma cells (including Y0 and NS0 cells), baby hamster kidney (BHK), Hela and Vera cells; (2) insect cells, for example, sf9, sf21 and Tn5; (3) plant cells, for example plants belonging to the genus Nicotiana (e.g. Nicotiana tabacum); (4) yeast cells, for example, those belonging to the genus Saccharomyces (e.g. Saccharomyces cerevisiae) or the genus Aspergillus (e.g. Aspergillus niger); (5) bacterial cells, for example Escherichia coli cells or Bacillus subtilis cells, etc.
  • mammalian cells for example, Chinese Hamster Ovary (CHO), COS, myeloma cells (including Y0 and NS0 cells), baby hamster kidney (B
  • binding affinity is generally determined using a standard assay, such as Scatchard analysis, or surface plasmon resonance technique (e.g. using BIACORE®).
  • an "antibody that binds to the same epitope” as a reference antibody refers to an antibody that blocks binding of the reference antibody to its antigen in a competition assay by 50% or more, and conversely, the reference antibody blocks binding of the antibody to its antigen in a competition assay by 50% or more.
  • an anti-BACEl antibody forming one of the bispecific or multispecific antibodies of the invention binds to the BACE1 epitope bound by YW412.8.31.
  • An exemplary competition assay is provided herein.
  • cytotoxic agent refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction.
  • Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu); chemotherapeutic agents or drugs (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal
  • an "effective amount" of an agent refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • Fc region herein is used to define a C-terminal region of an
  • immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native sequence Fc regions and variant Fc regions.
  • a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present.
  • numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 1991.
  • FcRn receptor or “FcRn” as used herein refers to an Fc receptor ("n" indicates neonatal) which is known to be involved in transfer of maternal IgGs to a fetus through the human or primate placenta, or yolk sac (rabbits) and to a neonate from the colostrum through the small intestine. It is also known that FcRn is involved in the
  • FcRn binding region or “FcRn receptor binding region” refers to that portion an an antibody which interacts with the FcRn receptor. Certain modifications in the FcRn binding region of an antibody increase the affinity of the antibody or fragment thereof, for the FcRn, and also increase the in vivo half-life of the molecule. Amino acid substitutions in one or more of the following amino acid positions 251 256, 285, 290, 308, 314, 385, 389, 428, 434 and 436 increases the interaction of the antibody with the FcRn receptor.
  • an “immunoconjugate” is an antibody conjugated to one or more heterologous molecule(s), including but not limited to a label or cytotoxic agent. Optionally such conjugation is via a linker.
  • a “linker” as used herein is a structure that covalently or non-covalently connects the anti-TfR antibody to heterologous molecule.
  • a linker is a peptide.
  • a linker is a chemical linker.
  • label is a marker coupled with the antibody herein and used for detection or imaging.
  • labels include: radiolabel, a fluorophore, a chromophore, or an affinity tag.
  • the label is a radiolabel used for medical imaging, for example tc99m or 1123, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine-123 again, iodine-131, indium-I l l, fluorine- 19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese, iron, etc.
  • NMR nuclear magnetic resonance
  • mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • domesticated animals e.g., cows, sheep, cats, dogs, and horses
  • primates e.g., humans and non-human primates such as monkeys
  • rabbits e.g., mice and rats
  • rodents e.g., mice and rats.
  • the individual or subject is a human.
  • an “isolated” antibody is one which has been separated from a component of its natural environment.
  • an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC) methods.
  • electrophoretic e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g., ion exchange or reverse phase HPLC
  • isolated nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment.
  • An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
  • isolated nucleic acid encoding an anti-TfR antibody refers to one ormore nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
  • Percent (%) amino acid sequence identity with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2.
  • the ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU 10087.
  • the ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code.
  • the ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
  • % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows:
  • pharmaceutical formulation refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • treatment refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • antibodies of the invention are used to delay development of a disease or to slow the progression of a disease.
  • variable region refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen.
  • the variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs).
  • FRs conserved framework regions
  • HVRs hypervariable regions
  • antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portolano et al., J.
  • vector refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked.
  • the term includes the vector as a self- replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors.”
  • the invention is based, in part, on anti-TfR antibodies that can be used to transport desired molecules across the BBB.
  • antibodies that bind to human TfR are provided.
  • antibodies that bind to both human TfR and primate TfR are provided.
  • Antibodies of the invention are useful, e.g., for the diagnosis or treatment of diseases affecting the brain and/or CNS.
  • the invention provides isolated antibodies that bind to TfR.
  • an anti-TfR antibody of the invention binds specifically to both human TfR and primate TfR.
  • an anti-TfR antibody of the invention does not inhibit binding of transferrin to the TfR.
  • an anti-TfR antibody of the invention binds to an apical domain of TfR.
  • an anti- TfR antibody of the invention binds to a non-apical domain of TfR.
  • the anti- TfR antibodies may be used to transport one or more conjugated imaging or therapeutic compounds across the BBB.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:32; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:33; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:34; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO:29; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 30; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 31.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 7A4, as shown in Figure 3 A and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:37; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:38; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:39; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO:35; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:30; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:36.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 8A2, as shown in Figure 3 A and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 32; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:40; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 34; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO: 35; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 30; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 36.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 15D2, as shown in Figure 3 A and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 37; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 43; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 44; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO: 41 ; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 30; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 42.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 10D 11 , as shown in Figure 3 A and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 32; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 33; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 34; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO: 29; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 30; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:31.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 7B10, as shown in Figure 3 A and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:53; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:54; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:55; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO: 50; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 1; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:52.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 15G11, as shown in Figure 3B and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:53; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:58; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:59; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO: 6; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:57; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:52.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 16G5, as shown in Figure 3B and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:53; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:63; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:55; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO: 60; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:61; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:62.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 13C3, as shown in Figure 3B and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:53; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:65; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:55; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO: 60; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:64; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:52.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 16G4, as shown in Figure 3B and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:74; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:75; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:76; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO:71; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO : 72; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO : 73.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 16F6, as shown in Figure 3C and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:80; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:81; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:82; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO: 77; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:78; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:79.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 7G7, as shown in Figure 3D and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 80; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 83; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:84; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO:77; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:78; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:79.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 4C2, as shown in Figure 3D and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:88; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:89; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:90; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO:85; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:86; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:87.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone IB 12, as shown in Figure 3D and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:94; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:95; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:96; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO:91; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 92; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 93.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 13D4, as shown in Figure 3D and Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO:32; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:33; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:34; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO:29; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 30; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 127.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the antibody is clone 7A4.vl5, as shown in Figure 4B and Table 4.
  • the clones above fall into four complementation groups, with sequence similarity within the HVRs. As shown in Table 3, consensus sequences are readily derivable from the provided antibody sequences for each HVR. As one nonlimiting example, the class I antibody consensus HVRs are as follows:
  • HVR-Ll Arg-Ala-Ser-Glu-Ser-Val-Asp-[Ser or Asp]-Tyr-Gly-[Asn or Pro]-Ser-Phe- Met-His (SEQ ID NO: 45);
  • HVR-L2 Arg-Ala-Ser-Asn-Leu-Glu-Ser (SEQ ID NO: 30);
  • HVR-L3 Gln-[Gln or His]-Ser-Asn-Glu-[Ala, Gly or Asp]-Pro-Pro-Thr (SEQ ID NO:
  • HVR-H1 Asp-Tyr-[Ala or Gly]-Met-His (SEQ ID NO: 47);
  • HVR-H2 [Gly or Val]-Ile-Ser-[Thr, Phe or Pro]-Tyr-[Phe or Ser]-Gly-[Arg or Lys]- Thr-Asn-Tyr-[Asn or Ser]-Gln-[Lys or Asn]-Phe-[Lys or Met]-Gly (SEQ ID NO: 48);
  • HVR-H3 Gly-Leu-Ser-Gly-Asn-[Tyr or Phe]-Val-[Met or Val]-Asp-[Tyr or Phe] (SEQ ID NO: 49). (see Table 4). The consensus sequences for class II and rv are also provided in Table 3.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO:47; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:48; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:49; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO:45; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:30; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:46.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO:53; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:69; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:70; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO: 66; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:67; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:68.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the invention provides an anti-TfR antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-Hl comprising the amino acid sequence of SEQ ID NO: 100; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 101 ; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 102; (d) HVR-Ll comprising the amino acid sequence of SEQ ID NO: 97; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO:98; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO:99.
  • the antibody comprises all six of the above-recited HVR sequences.
  • the invention provides an antibody comprising at least one, at least two, or all three VH HVR sequences of any of the antibodies described above.
  • the antibody comprises the HVR-H3 sequence of any one of the antibodies described above.
  • the antibody comprises the HVR-H3 and HVR-L3 sequences of any one of the antibodies described above.
  • the antibody comprises the HVR- H3, HVR-L3 and HVR-H2 sequences of any one of the antibodies described above.
  • the antibody comprises the HVR-Hl , HVR-H2 and HVR-H3 sequences of any one of the antibodies described above.
  • the invention provides an antibody comprising at least one, at least two or all three VL HVR sequences of any of the antibodies described above.
  • the antibody comprises the HVR-Ll , HVR-L2, and HVR-L3 sequences of any one of the antibodies described above.
  • an antibody of the invention comprises (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from the HVR-Hl, HVR-H2, and HVR-H3 sequences of any one of the antibodies described above; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from the HVR- Ll, HVR-L2 and HVR-L3 sequences of any one of the antibodies described above.
  • an anti-TfR antibody is humanized.
  • an anti-TfR antibody comprises HVRs as in any of the above embodiments, and further comprises an acceptor human framework, e.g. a human immunoglobulin framework or a human consensus framework.
  • an anti-TfR antibody comprises HVRs as in any of the above embodiments, and further comprises a VH or VL comprising one or more amino acid substitutions in one or more FR regions.
  • Applicants performed alanine scanning on certain antibodies selected from those above, and determined that similar or improved binding was obtained despite amino acid modifications at selected FR positions.
  • positions 58 and 68 of the light chain FR3, position 24 in the heavy chain FR1 and position 71 in the heavy chain FR3 could be modified as shown in Figures 6-1 and 6-2 and the resulting antibody still retained specificity and strong binding affinity for human/primate TfR.
  • positions 43 and 44 of the light chain FR2 and positions 71 and 78 of the heavy chain FR3 could be modified as shown in Figures 6- 1 and 6-2 and the resulting antibody still retained specificity and strong binding affinity for human/primate TfR.
  • an anti-TfR antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of any one of SEQ ID Os: 7-10, 15-18, 20, 25-28, 108, 114, 120 and 126.
  • VH heavy chain variable domain
  • a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-TfR antibody comprising that sequence retains the ability to bind to TfR.
  • the anti-TfR antibody comprises the VH sequence of any one of SEQ ID NOs: 7-10, 15-18, 20 25-28, 108, 114, 120 and 126, including post-translational modifications of that sequence.
  • the VH for a particular antibody comprises one, two or three HVRs selected from: the HVRs set forth above and in Table 3 or 4 for that particular antibody.
  • VH sequences for the antibodies of the invention are shown in Figures 3 and 4 herein.
  • an anti-TfR antibody comprising a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of any one of SEQ ID NOs:4-6, 11-14, 19, 21-24, 105, 111, 117, and 123.
  • VL light chain variable domain
  • a VL sequence having at least 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-TfR antibody comprising that sequence retains the ability to bind to TfR.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in any one of SEQ ID NOs:4-6, 11-14, 19, 21-24, 105, 111, 117, and 123 .
  • the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs).
  • the anti-TfR antibody comprises the VL sequence in any of SEQ ID NOs:4-6, 11-14, 19, 21-24, 105, 111, 117, and 123, including post-translational modifications of that sequence.
  • the VL comprises one, two or three HVRs selected from the HVRs set forth above and in Table 4 or 5 for that particular antibody. VL sequences for the antibodies of the invention are shown in Figures 3 and 4 herein..
  • an anti-TfR antibody comprising a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • the antibody comprises the VL and VH sequences, respectively, in SEQ ID NOs: 4 and 7; 5 and 8; 5 and 9; 6 and 10; 4 and 7; 11 and 15; 12 and 16; 13 and 17; 14 and 18; 19 and 20; 21 and 25; 22 and 26; 23 and 27; 24 and 28; 105 and 108; 111 and 114; 117 and 120; and 123 and 126 , including post-translational modifications of those sequences.
  • the invention provides an antibody that binds to the same epitope as an anti-TfR antibody provided herein.
  • an antibody is provided that binds to the same epitope as an anti-TfR antibody comprising VL and VH sequences, respectively, of SEQ ID NOs: 4 and 7; 5 and 8; 5 and 9; 6 and 10; 4 and 7; 11 and 15; 12 and 16; 13 and 17; 14 and 18; 19 and 20; 21 and 25; 22 and 26; 23 and 27; 24 and 28; 105 and 108; 111 and 114; 117 and 120; or 123 and 126.
  • the antibody competes with any of the antibodies in Class I (ie., clones 7A4, 8A2, 15D2, 10D11, or 7B10, or affmity- matured versions of any of those antibodies) for binding to TfR.
  • the antibody competes with any of the antibodies in Class II (ie, clones 15G1 1, 16G5, 13C3 or 16G, or affinity-matured versions of any of those antibodies) for binding to TfR.
  • the antibody competes with clone 16F6 or affinity-matured versions thereof for binding to TfR.
  • the antibody competes with any of the antibodies in Class IV (ie, clones 7G7, 4C2, IB 12 or 13D4, or affinity matured versions thereof) for binding to TfR.
  • an anti-TfR antibody is a monoclonal antibody, including a chimeric, humanized or human antibody.
  • an anti-TfR antibody is an antibody fragment, e.g., a Fv, Fab, Fab', scFv, diabody, or F(ab') 2 fragment.
  • the antibody is a full length antibody, e.g., an intact IgGl , IgG2, IgG3, or IgG4 antibody or other antibody class or isotype as defined herein.
  • an anti-TfR antibody may incorporate any of the features, singly or in combination, as described in Sections 1-7 below:
  • an antibody provided herein has a dissociation constant (Kd) of ⁇ ⁇ ⁇ , ⁇ ⁇ ⁇ , ⁇ ⁇ ⁇ , ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g. 10 ⁇ 8 M or less, e.g. from 10 ⁇ 8 M to 10 ⁇ 13 M, e.g., from 10 "9 M to 10 ⁇ 13 M).
  • Kd dissociation constant
  • a "low affinity" anti-TfR antibody of the invention is selected, based, e.g., on the results in Example 5 and in Atwal et al., Sci. Transl. Med. 3, 84ra43 (2011) and Yu et al, Sci. Transl. Med. 25 May 201 1 : Vol. 3, Issue 84, p.
  • the antibody has an affinity for human or primate TfR from about 5nM, or from about 20 nM, or from about 100 nM, to about 50 ⁇ , or to about 30 ⁇ , or to about 10 ⁇ , or to about 1 ⁇ , or to about 500 nM.
  • the affinity may be in the range from about 5 nM to about 50 ⁇ , or in the range from about 20 nM to about 30 ⁇ , or in the range from about 30 nM to about 30 ⁇ , or in the range from about 50 nM to about 1 ⁇ , or in the range from about 100 nM to about 500 nM, e.g. as measured by Scatchard analysis or BIACORE®.
  • the antibody has a dissociation half-life from TfR of less than 1 minute, less than 2 minutes, less than 3 minutes, less than four minutes, less than 5 minutes, or less than 10 minutes to about 20 minutes, or to about 30 minutes, as measured by competition binding analysis or BIACORE®.
  • the invention provides a method of making an antibody useful for transporting a neurological disorder drug across the blood-brain barrier comprising selecting an antibody from a panel of antibodies against TfR because it has an affinity for TfR which is in the range from about 5nM, or from about 20 nM, or from about 100 nM, to about 50 ⁇ , or to about 30 ⁇ , or to about 10 ⁇ , or to about 1 ⁇ , or to about 500 mM.
  • the affinity may be in the range from about 5 nM to about 50 ⁇ , or in the range from about 20 nM to about 30 ⁇ , or in the range from about 30 nM to about 30 ⁇ , or in the range from about 50 nM to about 1 ⁇ , or in the range from about 100 nM to about 500 nM, e.g. as measured by Scatchard analysis or BIACORE®.
  • a low affinity antibody of the invention specific for TfR conjugated to anti-BACEl had a Kd for TfR as measured by BIACORE of about 30 nM.
  • a low affinity antibody of the invention specific for TfR conjugated to BACE1 had a Kd for TfR as measured by BIACORE of about 600 nM.
  • BACE 1 had a Kd for TfR as measured by BIACORE of about 20 ⁇ .
  • a low affinity antibody of the invention specific for TfR conjugated to BACE 1 had a Kd for TfR as measured by BIACORE of about 30 ⁇ .
  • Kd is measured by a radiolabeled antigen binding assay (RIA).
  • RIA radiolabeled antigen binding assay
  • an RIA is performed with the Fab version of an antibody of interest and its antigen.
  • solution binding affinity of Fabs for antigen is measured by
  • MICROTITER multi-well plates (Thermo Scientific) are coated overnight with 5 ⁇ 3 ⁇ 4 / ⁇ 1 of a capturing anti-Fab antibody (Cappel Labs) in 50 mM sodium carbonate (pH 9.6), and subsequently blocked with 2% (w/v) bovine serum albumin in PBS for two to five hours at room temperature (approximately 23°C).
  • a non-adsorbent plate (Nunc #269620)
  • 100 pM or 26 pM [ 125 I]-anti gen are mixed with serial dilutions of a Fab of interest (e.g., consistent with assessment of the anti-VEGF antibody, Fab-12, in Presta et al., Cancer Res.
  • the Fab of interest is then incubated overnight; however, the incubation may continue for a longer period (e.g., about 65 hours) to ensure that equilibrium is reached. Thereafter, the mixtures are transferred to the capture plate for incubation at room temperature (e.g., for one hour). The solution is then removed and the plate washed eight times with 0.1% polysorbate 20 (TWEEN-20 ® ) in PBS. When the plates have dried, 150 ⁇ /well of scintillant
  • the RJA is a Scatchard analysis.
  • the anti-TfR antibody of interest can be iodinated using the lactoperoxidase method (Bennett and Horuk, Methods in Enzymology 288 pg.134-148 (1997)).
  • a radiolabeled anti-TfR antibody is purified from free
  • 125 I-Na by gel filtration using a NAP-5 column and its specific activity measured.
  • Competition reaction mixtures 50 ⁇ L containing a fixed concentration of iodinated antibody and decreasing concentrations of serially diluted unlabeled antibody are placed into 96-well plates.
  • Cells transiently expressing TfR are cultured in growth media, consisting of Dulbecco's modified eagle's medium (DMEM) (Genentech) supplemented with 10% FBS, 2 mM L-glutamine and 1 x penicillin-streptomycin at 37°C in 5% C0 2 .
  • DMEM Dulbecco's modified eagle's medium
  • Bind Cells are detached from the dishes using Sigma Cell Dissociation Solution and washed with binding buffer (DMEM with 1% bovine serum albumin, 50 mM HEPES, pH 7.2, and 0.2% sodium azide). The washed cells are added at an approximate density of 200,000 cells in 0.2 mL of binding buffer to the 96-well plates containing the 50- ⁇ competition reaction mixtures. The final concentration of the unlabeled antibody in the competition reaction with cells is varied, starting at 1000 nM and then decreasing by 1 :2 fold dilution for 10 concentrations and including a zero-added, buffer-only sample. Competition reactions with cells for each concentration of unlabeled antibody are assayed in triplicate. Competition reactions with cells are incubated for 2 hours at room temperature.
  • binding buffer DMEM with 1% bovine serum albumin, 50 mM HEPES, pH 7.2, and 0.2% sodium azide.
  • the washed cells are added at an approximate density of 200,000 cells in 0.2 mL of binding buffer to the 96
  • the competition reactions are transferred to a filter plate and washed four times with binding buffer to separate free from bound iodinated antibody.
  • the filters are counted by gamma counter and the binding data are evaluated using the fitting algorithm of Munson and Rodbard (1980) to determine the binding affinity of the antibody.
  • CM5 carboxymethylated dextran biosensor chips
  • EDC N-ethyl-N'- (3-dimethylaminopropyl)-carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • Anti-human Fc antibody was diluted with 10 mM sodium acetate, pH 4.0, to 50 ⁇ g/ml before injection at a flow rate of 5 ⁇ /minute to achieve approximately 10000 response units (RU) of coupled protein. Following the injection of antibody, 1 M ethanolamine was injected to block unreacted groups. For kinetics measurements, monospecific or multispecific anti-TfR antibody variants were injected in HBS-P to reach about 220 RU, then two-fold serial dilutions of MuTfR-His (0.61 nM to 157 nM) were injected in HBS-P at 25°C at a flow rate of approximately 30 ⁇ /min.
  • Kd is measured using surface plasmon resonance assays with a BIACORE®-2000 device (BIAcore, Inc., Piscataway, NJ) at 25°C using anti- human Fc kit (BiAcore Inc., Piscataway, NJ).
  • CM5 carboxymethylated dextran biosensor chips
  • EDC N-ethyl-N'- (3-dimethylaminopropyl)- carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • Anti-human Fc antibody is diluted with 10 mM sodium acetate, pH 4.0, to 50 ⁇ g/ml before injection at a flow rate of 5 ⁇ /minute to achieve approximately 10000 response units (RU) of coupled protein. Following the injection of antibody, 1 M ethanolamine is injected to block unreacted groups.
  • anti-TfR antibody variants are injected in HBS-P to reach about 220 RU, then two-fold serial dilutions of TfR-His (0.61 nM to 157 nM) are injected in HBS-P at 25°C at a flow rate of approximately 30 ⁇ /min.
  • association rates (kon) and dissociation rates (koff) are calculated using a simple one-to-one Langmuir binding model (BIACORE ® Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams.
  • the equilibrium dissociation constant (Kd) is calculated as the ratio koff/kon. See, e.g., Chen et al, J. Mol. Biol. 293 :865-881 (1999).
  • IC50 for a given compound
  • a common approach is to perform a competition binding assay, such as that described herein.
  • a high IC50 indicates that more of the antibody is required to inhibit binding of the known ligand, and thus that the antibody's affinity for that ligand is relatively low.
  • a low IC 0 indicates that less of the antibody is required to inhibit binding of the known ligand, and thus that the antibody's affinity for that ligand is relatively high.
  • An exemplary competitive ELISA assay to measure IC50 is one in which increasing concentrations of anti-Tf or anti-TfR/brain antigen (i.e., anti-TfR/BACE 1 , anti-TfR Abeta and the like) variant antibodies are used to compete against a biotinylated known anti-TfR antibody for binding to TfR.
  • the anti-TfR competition ELISA was performed in Maxisorp plates (Neptune, N.J.) coated with 2.5 ⁇ g/ml of purified murine TfR extracellular domain in PBS at 4°C overnight. Plates were washed with PBS/0.05% Tween 20 and blocked using Superblock blocking buffer in PBS (Thermo Scientific, Hudson, NH).
  • an antibody provided herein is an antibody fragment.
  • Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab')2, Fv, and scFv fragments, and other fragments described below.
  • Fab fragment antigen binding fragment
  • Fab' fragment antigen binding fragment
  • Fab'-SH fragment antigen binding fragment
  • F(ab')2 fragment antigen binding fragment
  • scFv fragments fragment antigen binding fragments
  • other fragments described below For a review of certain antibody fragments, see Hudson et al. Nat. Med. 9:129-134 (2003).
  • scFv fragments see, e.g., Pluckthun, in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., (Springer- Verlag, New York), pp. 269-315 (1994); see also WO 93/16185; and U.S. Patent Nos. 5,571,894 and 5,587,458.
  • Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson et al, Nat. Med. 9:129- 134 (2003); and Hollinger et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993). Triabodies and tetrabodies are also described in Hudson et al, Nat. Med. 9: 129-134 (2003).
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U.S. Patent No. 6,248,516 Bl).
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
  • recombinant host cells e.g. E. coli or phage
  • an antibody provided herein is a chimeric antibody.
  • Certain chimeric antibodies are described, e.g., in U.S. Patent No. 4,816,567; and Morrison et al, Proc. Natl. Acad. Sci. USA, 81 :68 1-6855 (1984)).
  • a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region.
  • Chimeric antibodies of interest herein include "primatized" antibodies comprising variable domain antigen-binding sequences derived from a non-human primate (e.g.
  • a chimeric antibody is a "class switched" antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.
  • a chimeric antibody is a humanized antibody.
  • a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • a humanized antibody comprises one or more variable domains in which HVRs, e.g., CDRs, (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences.
  • HVRs e.g., CDRs, (or portions thereof) are derived from a non-human antibody
  • FRs or portions thereof
  • a humanized antibody optionally will also comprise at least a portion of a human constant region.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the HVR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • a non-human antibody e.g., the antibody from which the HVR residues are derived
  • Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the "best-fit" method (see, e.g., Sims et al. J. Immunol. 151 :2296 (1993)); framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light or heavy chain variable regions (see, e.g., Carter et al. Proc. Natl. Acad. Sci. USA, 89:4285 (1992); and Presta et al. J. Immunol , 151 :2623 (1993)); human mature (somatically mutated) framework regions or human germline framework regions (see, e.g., Almagro and Fransson, Front. Biosci.
  • an antibody provided herein is a human antibody.
  • Human antibodies can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, Curr. Opin. Pharmacol. 5 : 368-74 (2001) and Lonberg, Curr. Opin. Immunol. 20:450-459 (2008).
  • Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge. Such animals typically contain all or a portion of the human immunoglobulin loci, which replace the endogenous
  • immunoglobulin loci or which are present extrachromosomally or integrated randomly into the animal's chromosomes. In such transgenic mice, the endogenous immunoglobulin loci have generally been inactivated.
  • endogenous immunoglobulin loci have generally been inactivated.
  • Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., KozborJ. Immunol, 133: 3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987); and Boerner et al., J. Immunol., 147: 86 (1991).) Human antibodies generated via human B-ceil hybridoma technology are also described in Li et al., Proc. Natl. Acad. Sci. USA, 103:3557-3562 (2006).
  • Additional methods include those described, for example, in U.S. Patent No. 7,189,826 (describing production of monoclonal human IgM antibodies from hybridoma cell lines) and ⁇ , ⁇ Mianyixue, 26(4):265-268 (2006) (describing human-human hybridomas).
  • Human hybridoma technology Trioma technology
  • Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below.
  • Antibodies of the invention may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. For example, a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, e.g., in
  • repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PCR) and recombined randomly in phage libraries, which can then be screened for antigen-binding phage as described in Winter et al., Ann. Rev. Immunol, 12: 433-455 (1994).
  • Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments.
  • naive repertoire can be cloned (e.g., from human) to provide a single source of antibodies to a wide range of non-self and also self antigens without any immunization as described by Griffiths et al., EMBO J, 12: 725-734 (1993).
  • naive libraries can also be made synthetically by cloning unrearranged V-gene segments from stem cells, and using PCR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom and Winter, J. Mol. Biol, 227: 381-388 (1992).
  • Patent publications describing human antibody phage libraries include, for example: US Patent No. 5,750,373, and US Patent Publication Nos. 2005/0079574, 2005/0119455, 2005/0266000, 2007/0117126, 2007/0160598, 2007/0237764, 2007/0292936, and 2009/0002360.
  • Antibodies or antibody fragments isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.
  • an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody.
  • Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites. In certain embodiments, one of the binding specificities is for TfR and the other is for any other antigen.
  • bispecific antibodies may bind to two different epitopes of TfR. Bispecific antibodies may also be used to localize cytotoxic agents to cells which express TfR. Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
  • Multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein and Cuello, Nature 305 : 537 (1983)), WO 93/08829, and Traunecker et al., EMBO J. 10: 3655 (1991)), and "knob-in-hole” engineering (see, e.g., U.S. Patent No. 5,731 ,168). Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross- linking two or more antibodies or fragments (see, e.g., US Patent No.
  • the antibody or fragment herein also includes a "Dual Acting FAb” or “DAF” comprising an antigen binding site that binds to TfR as well as another, different antigen (see, US 2008/0069820, for example).
  • the "coupling" is achieved by generating a multispecific antibody (e.g. a bispecific antibody).
  • Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different antigens or epitopes.
  • the multispecific antibody comprises a first antigen binding site which binds the TfR and a second antigen binding site which binds a brain antigen, such as beta-secretase 1 (BACE1) or Abeta, and the other brain antigens disclosed herein.
  • BACE1 beta-secretase 1
  • An exemplary brain antigen bound by such multispecific/bispecific antibody is BACE 1
  • an exemplary antibody binding thereto is the YW412.8.31 antibody in Figs. 16A-B herein.
  • the brain antigen is Abeta, exemplary such antibodies being described in WO2007068412, WO2008011348, WO20080156622, and WO2008156621, expressly incorporated herein by reference, with an exemplary Abeta antibody comprising the IgG4 MABT5102A antibody comprising the heavy and light chain amino acid sequences in Figs. 11A and 1 IB, respectively.
  • Multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein and Cuello, Nature 305: 537 (1983)), WO 93/08829, and Traunecker et al, EMBOJ. 10: 3655 (1991)), and "knob-in-hole” engineering (see, e.g., U.S. Patent No. 5,731,168).
  • Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross- linking two or more antibodies or fragments (see, e.g., US Patent No.
  • Engineered antibodies with three or more functional antigen binding sites including
  • Optus antibodies or “dual-variable domain immunoglobulins” (DVDs) are also included herein (see, e.g. US 2006/0025576A1, and Wu et al. Nature Biotechnology (2007)). 7. Antibody Variants
  • amino acid sequence variants of the antibodies provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding.
  • antibody variants having one or more amino acid substitutions are provided.
  • Sites of interest for substitutional mutagenesis include the HVRs and FRs.
  • amino acid side chain classes Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased
  • Amino acids may be grouped according to common side-chain properties:
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody).
  • a parent antibody e.g. a humanized or human antibody
  • the resulting variant(s) selected for further study will have modifications (e.g., improvements) in certain biological properties (e.g., increased affinity, reduced immunogenicity) relative to the parent antibody and/or will have substantially retained certain biological properties of the parent antibody.
  • An exemplary substitutional variant is an affinity matured antibody, which may be conveniently generated, e.g., using phage display-based affinity maturation techniques such as those described herein. Briefly, one or more HVR residues are mutated and the variant antibodies displayed on phage and screened for a particular biological activity (e.g. binding affinity).
  • Alterations may be made in HVRs, e.g., to improve antibody affinity. Such alterations may be made in HVR "hotspots," i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol. 207: 179-196 (2008)), and/or residues that contact antigen, with the resulting variant VH or VL being tested for binding affinity.
  • HVR "hotspots” i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol. 207: 179-196 (2008)), and/or residues that contact antigen, with the resulting variant VH or VL being tested for binding affinity.
  • Affinity maturation by constructing and reselecting from secondary libraries has been described, e.g., in Hoogenboom et al.
  • affinity maturation diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oligonucleotide-directed mutagenesis).
  • a secondary library is then created. The library is then screened to identify any antibody variants with the desired affinity.
  • Another method to introduce diversity involves HVR-directed approaches, in which several HVR residues (e.g., 4-6 residues at a time) are randomized. HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted.
  • substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen.
  • conservative alterations e.g., conservative substitutions as provided herein
  • Such alterations may, for example, be outside of antigen contacting residues in the HVRs.
  • each HVR either is unaltered, or contains no more than one, two or three amino acid substitutions.
  • a useful method for identification of residues or regions of an antibody that may be targeted for mutagenesis is called "alanine scanning mutagenesis" as described by Cunningham and Wells (1989) Science, 244:1081-1085.
  • a residue or group of target residues e.g., charged residues such as arg, asp, his, lys, and glu
  • a neutral or negatively charged amino acid e.g., alanine or polyalanine
  • a crystal structure of an antigen-antibody complex to identify contact points between the antibody and antigen.
  • Such contact residues and neighboring residues may be targeted or eliminated as candidates for substitution.
  • Variants may be screened to determine whether they contain the desired properties.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an antibody with an N-terminal methionyl residue.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g. for ADEPT) or a polypeptide which increases the serum half-life of the antibody.
  • an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated.
  • Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
  • the carbohydrate attached thereto may be altered.
  • Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright et al. TIBTECH 15 :26-32 (1997).
  • oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the "stem" of the biantennary oligosaccharide structure.
  • modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.
  • antibody variants having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region.
  • the amount of fucose in such antibody may be from 1% to 80%, from 1% to 65%, from 5% to 65% or from 20% to 40%.
  • the amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example.
  • Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ⁇ 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies.
  • Such fucosylation variants may have improved ADCC function. See, e.g., US Patent Publication Nos. US 2003/0157108 (Presta, L.); US 2004/0093621 ( yowa Hakko Kogyo Co., Ltd). Examples of publications related to "defucosylated” or "fucose-deficient" antibody variants include: US 2003/0157108; WO 2000/61739; WO 2001/29246; US
  • Examples of cell lines capable of producing defucosylated antibodies include Lecl3 CHO cells deficient in protein fucosylation (Ripka et al. Arch. Biochem. Biophys. 249:533-545 (1986); US Pat Appl No US 2003/0157108 Al , Presta, L; and WO 2004/056312 Al, Adams et al, especially at Example 11), and knockout cell lines, such as alpha- 1 ,6-fucosyltransferase gene, FUT8, knockout CHO cells (see, e.g., Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004); Kanda, Y. et al, Biotechnol. Bioeng., 94(4):680-688 (2006); and WO2003/08 107).
  • Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/01 1878 (Jean-Mairet et al.); US Patent No. 6,602,684 (Umana et al); and US 2005/0123546 (Umana et al.). Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided.
  • Such antibody variants may have improved CDC function.
  • Such antibody variants are described, e.g., in WO 1997/30087 (Patel et al); WO 1998/58964 (Raju, S.); and WO 1999/22764 (Raju, S.).
  • one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant.
  • the Fc region variant may comprise a human Fc region sequence ⁇ e.g., a human IgGl , IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification ⁇ e.g. a substitution) at one or more amino acid positions.
  • the invention contemplates an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious.
  • In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities.
  • Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability.
  • non-radioactive assays methods may be employed (see, for example, ACTITM non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, CA; and CytoTox
  • PBMC peripheral blood mononuclear cells
  • NK Natural Killer
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al. Proc. Nat ⁇ Acad. Sci. USA 95:652-656 (1998).
  • Clq binding assays may also be carried out to confirm that the antibody is unable to bind Clq and hence lacks CDC activity. See, e.g., Clq and C3c binding ELISA in
  • a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J. Immunol. Methods 202: 163 (1996);
  • FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, S.B. et al., Intl. Immunol.
  • Non-limiting examples of antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Patent No. 6,737,056).
  • Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called "DANA" Fc mutant with substitution of residues 265 and 297 to alanine (US Patent No. 7,332,581).
  • an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298, 333, and/or
  • alterations are made in the Fc region that result in altered (i.e., either improved or diminished) Clq binding and/or Complement Dependent Cytotoxicity
  • Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US Patent No. 7,371,826).
  • cysteine engineered antibodies e.g., "thioMAbs”
  • one or more residues of an antibody are substituted with cysteine residues.
  • the substituted residues occur at accessible sites of the antibody.
  • reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein.
  • any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; Al 18 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region.
  • Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No.
  • an antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available.
  • the moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers.
  • water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3- dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)polyethylene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols (e.g., gly
  • Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the number of polymers attached to the antibody may vary, and if more than one polymer are attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
  • conjugates of an antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided.
  • the nonproteinaceous moiety is a carbon nanotube (Kam et al., Proc. Natl. Acad. Sci. USA 102: 11600-11605 (2005)).
  • the radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody-nonproteinaceous moiety are killed.
  • Antibodies may be produced using recombinant methods and compositions, e.g., as described in U.S. Patent No. 4,816,567.
  • isolated nucleic acid encoding an anti-TfR antibody described herein is provided.
  • Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody).
  • one or more vectors e.g., expression vectors
  • a host cell comprising such nucleic acid is provided.
  • a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody.
  • the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NS0, Sp20 cell).
  • a method of making an anti-Tf antibody comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
  • nucleic acid encoding an antibody is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • nucleic acid may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
  • Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein.
  • antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed.
  • U.S. Patent Nos. 5,648,237, 5,789,199, and 5,840,523. See also Charlton, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ, 2003), pp. 245-254, describing expression of antibody fragments in E. coli.
  • the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been "humanized,” resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, Nat. Biotech. 22:1409-1414 (2004), and Li et al, Nat. Biotech. 24:210-215 (2006).
  • Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures can also be utilized as hosts. See, e.g., US Patent Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIESTM technology for producing antibodies in transgenic plants).
  • Vertebrate cells may also be used as hosts.
  • mammalian cell lines that are adapted to grow in suspension may be useful.
  • useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BH ); mouse Sertoli cells (TM4 cells as described, e.g., in Mather, Biol. Reprod.
  • monkey kidney cells (CV1); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3 A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather et al., Annals N. Y. Acad. Sci. 383:44-68 (1982); MRC 5 cells; and FS4 cells.
  • Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR " CHO cells (Urlaub et al., Proc. Natl. Acad. Sci.
  • Anti-TfR antibodies provided herein may be identified, screened for, or characterized for their physical/chemical properties and/or biological activities by various assays known in the art.
  • One such assay is an enzyme linked immunosorbent assay (ELISA) for confirming an ability to bind to human TfR (and brain antigen).
  • ELISA enzyme linked immunosorbent assay
  • plates coated with antigen e.g. recombinant TfR
  • binding of the antibody to the antigen of interest is determined.
  • an antibody of the invention is tested for its antigen binding activity, e.g., by known methods such as ELISA, Western blot, etc.
  • competition assays may be used to identify an antibody that competes with any of the antibodies of the invention for binding to TfR.
  • a competing antibody binds to the same epitope (e.g., a linear or a conformational epitope) that is bound by any of the antibodies of the invention, more specifically, any of the epitopes specifically bound by antibodies in class I, class II, class III or class IV as described herein (see, e.g., Example 1 and Table 4.
  • Detailed exemplary methods for mapping an epitope to which an antibody binds are provided in Morris (1996) "Epitope Mapping Protocols," in Methods in Molecular Biology vol. 66 (Humana Press, Totowa, NJ).
  • immobilized TfR is incubated in a solution comprising a first labeled antibody that binds to TfR (e.g., one or more of the antibodies disclosed herein) and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to TfR.
  • the second antibody may be present in a hybridoma supernatant.
  • immobilized TfR is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After incubation under conditions permissive for binding of the first antibody to TfR, excess unbound antibody is removed, and the amount of label associated with immobilized TfR is measured.
  • assays are provided for identifying anti-TfR antibodies thereof having biological activity.
  • Biological activity may include, e.g., transporting a compound associated with/conjugated to the antibody across the BBB into the brain and/or CNS.
  • Antibodies having such biological activity in vivo and/or in vitro are also provided.
  • an antibody of the invention is tested for such biological activity.
  • the invention also provides immunoconjugates comprising an anti-TfR antibody herein conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • cytotoxic agents such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • the anti-TfR antibody herein is coupled with a neurological disorder drug, a chemotherapeutic agent and/or an imaging agent in order to more efficiently transport the drug, chemotherapeutic agent and/or the imaging agent across the BBB.
  • Covalent conjugation can either be direct or via a linker.
  • direct conjugation is by construction of a protein fusion (i.e., by genetic fusion of the two genes encoding the anti-TfR antibody and e.g., the neurological disorder drug and expression as a single protein).
  • direct conjugation is by formation of a covalent bond between a reactive group on one of the two portions of the anti-TfR antibody and a
  • direct conjugation is by modification (i.e., genetic modification) of one of the two molecules to be conjugated to include a reactive group (as nonlimiting examples, a sulfhydryl group or a carboxyl group) that forms a covalent attachment to the other molecule to be conjugated under appropriate conditions.
  • a reactive group as nonlimiting examples, a sulfhydryl group or a carboxyl group
  • a molecule i.e., an amino acid
  • a desired reactive group i.e., a cysteine residue
  • Non-covalent conjugation can be by any nonconvalent attachment means, including hydrophobic bonds, ionic bonds, electrostatic interactions, and the like, as will be readily understood by one of ordinary skill in the art.
  • an anti- TfR antibody and a neurological drug may be conjugated using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl- 4-( -maleimidomethyl) cyclohexane- 1 -carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HQ), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p- diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6
  • SPDP N-succinimidyl-3-(2-pyr
  • a ricin immunotoxin can be prepared as described in Vitetta et al., Science 238: 1098 (1987).
  • Carbon- 14-labeled l-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See
  • Peptide linkers comprised of from one to twenty amino acids joined by peptide bonds, may also be used.
  • the amino acids are selected from the twenty naturally-occurring amino acids.
  • one or more of the amino acids are selected from glycine, alanine, proline, asparagine, glutamine and lysine.
  • the linker may be a "cleavable linker" facilitating release of the neurological drug upon delivery to the brain.
  • an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari et al., Cancer Res. 52: 127-131 (1992); U.S. Patent No. 5,208,020) may be used.
  • the invention herein expressly contemplates, but is not limited to, conjugates prepared with cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC- SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB
  • cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC- SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MB
  • an immunoconjugate is an antibody-drug conjugate (ADC) in which an antibody is conjugated to one or more drugs, including but not limited to a maytansinoid (see U.S. Patent Nos. 5,208,020, 5,416,064 and European Patent EP 0 425 235 Bl); an auristatin such as monomethylauristatin drug moieties DE and DF (MMAE and MMAF) (see U.S. Patent Nos. 5,635,483 and 5,780,588, and 7,498,298); a dolastatin; a calicheamicin or derivative thereof (see U.S. Patent Nos. 5,712,374, 5,714,586, 5,739,116, 5,767,285, 5,770,701, 5,770,710, 5,773,001, and 5,877,296; Hinman et al, Cancer Res.
  • ADC antibody-drug conjugate
  • an immunoconjugate comprises an antibody as described herein conjugated to an enzymatically active toxin or fragment thereof, including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha- sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • an enzymatically active toxin or fragment thereof including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain
  • an immunoconjugate comprises an antibody as described herein conjugated to a radioactive atom to form a radioconjugate.
  • a radioactive isotope are available for the production of radioconjugates. Examples include At 211 , 1131 , 1125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu.
  • the radioconjugate When used for detection, it may comprise a radioactive atom for scintigraphic studies, for example tc99m or 1123, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine- 123 again, iodine- 131, indium-I l l, fluorine-19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron.
  • NMR nuclear magnetic resonance
  • any of the anti-TfR antibodies provided herein is useful for detecting the presence of TfR in a biological sample.
  • the term "detecting" as used herein encompasses quantitative or qualitative detection.
  • a biological sample comprises a cell or tissue, such as blood (i.e., immature red blood cells), CSF, and BBB- containing tissue.
  • an anti-TfR antibody for use in a method of diagnosis or detection is provided.
  • a method of detecting the presence of TfR in a biological sample is provided.
  • the method comprises contacting the biological sample with an anti-TfR antibody as described herein under conditions permissive for binding of the anti-TfR antibody to TfR, and detecting whether a complex is formed between the anti- TfR antibody and TfR.
  • Such method may be an in vitro or in vivo method.
  • an anti-TfR antibody is used to select subjects eligible for therapy with an anti- TfR antibody, e.g. where TfR is a biomarker for selection of patients.
  • Exemplary disorders that may be diagnosed using an antibody of the invention includedisorders involving immature red blood cells, due to the fact that TfR is expressed in reticulocytes and is therefore detectable by any of the antibodies of the invention.
  • Such disorders include anemia and other disorders arising from reduced levels of reticulocytes, or congenital polycythemia or neoplastic polycythemia vera, where raised red blood cell counts due to hyperproliferation of, e.g., reticulocytes, results in thickening of blood and concomitant physiological symptoms. .
  • labeled anti-TfR antibodies are provided.
  • Labels include, but are not limited to, labels or moieties that are detected directly (such as fluorescent,
  • chromophoric, electron-dense, chemiluminescent, and radioactive labels as well as moieties, such as enzymes or ligands, that are detected indirectly, e.g., through an enzymatic reaction or
  • Exemplary labels include, but are not limited to, the radioisotopes P, 14 C, 125 1, H, and 1 1 I, fluorophores such as rare earth chelates or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, luceriferases, e.g., firefly luciferase and bacterial luciferase (U.S. Patent No.
  • luciferin 2,3-dihydrophthalazinediones
  • horseradish peroxidase HRP
  • alkaline phosphatase alkaline phosphatase
  • ⁇ -galactosidase glucoamylase
  • lysozyme saccharide oxidases, e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase
  • heterocyclic oxidases such as uricase and xanthine oxidase, coupled with an enzyme that employs hydrogen peroxide to oxidize a dye precursor such as HRP
  • HRP horseradish peroxidase
  • lactoperoxidase or microperoxidase, biotin/avidin, spin labels, bacteriophage labels, stable free radicals, and the like.
  • the intact antibody lacks effector function.
  • the inact antibody has reduced effector function.
  • the intact antibody is engineered to have reduced effector function.
  • the antibody is a Fab.
  • the antibody has one or more Fc mutations reducing or eliminating effector function.
  • the antibody has modified glycosylation due, e.g., to producing the antibody in a system lacking normal human glycosylation enzymes.
  • the Ig backbone is modified to one which naturally possesses limited or no effector function.
  • One such assay is an enzyme linked immunosorbent assay (ELISA) for confirming an ability to bind to human TfR (and brain antigen).
  • ELISA enzyme linked immunosorbent assay
  • plates coated with antigen e.g. recombinant TfR
  • binding of the antibody to the antigen of interest is determined.
  • Assays for evaluating uptake of systemically administered antibody and other biological activity of the antibody can be performed as disclosed in the examples or as known for the anti- CNS antigen antibody of interest.
  • assays are provided for identifying anti-TfR antibodies conjugated (either covalently or non-covalently) to anti-BACEl antibodies having biological activity.
  • Biological activity may include, e.g., inhibition of BACE1 aspartyl protease activity.
  • Antibodies having such biological activity in vivo and/or in vitro are also provided, e.g. as evaluated by homogeneous time-resolved fluorescence HTRF assay or a microfluidic capillary
  • MCE electrophoretic
  • compositions of an anti-TfR as described herein are prepared by mixing such antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington 's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Pharmaceutically acceptable carriers, excipients, or stabilizers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride;
  • hexamethonium chloride benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol;
  • polypeptides such as serum albumin, gelatin, or immunoglobulins
  • proteins such as serum albumin, gelatin, or immunoglobulins
  • hydrophilic polymers such as polyvinylpyrrolidone
  • amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine
  • monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g.
  • sHASEGP soluble neutral-active hyaluronidase glycoproteins
  • rHuPH20 HYLENEX ® , Baxter International, Inc.
  • a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
  • Exemplary lyophilized antibody formulations are described in US Patent No. 6,267,958.
  • Aqueous antibody formulations include those described in US Patent No. 6, 171,586 and WO2006/044908, the latter formulations including a histidine-acetate buffer.
  • the formulation herein may also contain more than one active ingredient as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
  • Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres,
  • microemulsions such techniques are disclosed in, for example, Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
  • active ingredients may be encapsulated in liposomes that are coupled to anti-TfPv antibodies described herein (see e.g., U.S. Patent Application Publication No.
  • sustained-release preparations may be prepared.
  • suitable examples of sustained- release preparations include semi-permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • Nonlimiting examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and y ethyl-L-glutamate copolymers of L-glutamic acid and y ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid.
  • LUPRON DEPOTTM injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate
  • poly-D-(-)-3-hydroxybutyric acid poly-D-(-)-3-hydroxybutyric acid.
  • the formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
  • an anti-TfR antibody for use as a medicament is provided.
  • the invention provides a method of transporting a therapeutic compound across the blood-brain barrier with reduced or eliminated impact on red blood cell populations comprising exposing the anti-TfR antibody coupled to a therapeutic compound (e.g. a multispecific antibody which binds both the TfR and a brain antigen) to the BBB such that the antibody transports the therapeutic compound coupled thereto across the BBB.
  • a therapeutic compound e.g. a multispecific antibody which binds both the TfR and a brain antigen
  • the invention provides a method of transporting a neurological disorder drug across the blood-brain barrier comprising exposing an anti-TfR antibody of the invention coupled to a brain disorder drug (e.g.
  • the BBB is in a mammal (e.g. a human), e.g. one which has a neurological disorder, including, without limitation: Alzheimer's disease (AD), stroke, dementia, muscular dystrophy (MD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), cystic fibrosis, Angelman's syndrome, Liddle syndrome, Parkinson's disease, Pick's disease, Paget's disease, cancer, traumatic brain injury, etc.
  • a mammal e.g. a human
  • a neurological disorder including, without limitation: Alzheimer's disease (AD), stroke, dementia, muscular dystrophy (MD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), cystic fibrosis, Angelman's syndrome, Liddle syndrome, Parkinson's disease, Pick's disease, Paget's disease, cancer, traumatic brain injury, etc.
  • the neurological disorder is selected from: a neuropathy, an amyloidosis, cancer (e.g. involving the CNS or brain), an ocular disease or disorder, a viral or microbial infection, inflammation (e.g. of the CNS or brain), ischemia, neurodegenerative disease, seizure, behavioral disorder, lysosomal storage disease, etc.
  • the antibodies of the invention are particularly suited to treatment of such neurological disorders due to their ability to transport one or more associated active ingredients/coupled therapeutic compounds across the BBB and into the CNS/brain where such disorders find their molecular, cellular, or viral/microbial basis.
  • Neuropathy disorders are diseases or abnormalities of the nervous system characterized by inappropriate or uncontrolled nerve signaling or lack thereof, and include, but are not limited to, chronic pain (including nociceptive pain), pain caused by an injury to body tissues, including cancer-related pain, neuropathic pain (pain caused by abnormalities in the nerves, spinal cord, or brain), and psychogenic pain (entirely or mostly related to a psychological disorder), headache, migraine, neuropathy, and symptoms and syndromes often accompanying such neuropathy disorders such as vertigo or nausea.
  • chronic pain including nociceptive pain
  • pain caused by an injury to body tissues including cancer-related pain
  • neuropathic pain pain caused by abnormalities in the nerves, spinal cord, or brain
  • psychogenic pain include, but are not limited to, headache, migraine, neuropathy, and symptoms and syndromes often accompanying such neuropathy disorders such as vertigo or nausea.
  • a neurological drug may be selected that is an analgesic including, but not limited to, a narcotic/opioid analgesic (i.e., morphine, fentanyl, hydrocodone, meperidine, methadone, oxymorphone, pentazocine, propoxyphene, tramadol, codeine and oxycodone), a nonsteroidal anti-inflammatory drug (NSAID) (i.e., ibuprofen, naproxen, diclofenac, diflunisal, etodolac, fenoprofen, flurbiprofen, indomethacin, ketorolac, mefenamic acid, meloxicam, nabumetone, oxaprozin, piroxicam, sulindac, and tolmetin), a corticosteroid (i.e., cortisone, prednisone, prednisolone, dexa
  • NSAID non
  • an anti-migraine agent i.e., sumatriptin, almotriptan, frovatriptan, sumatriptan, rizatriptan, eletriptan, zolmitriptan, dihydroergotamine, eletriptan and ergotamine
  • an anti-migraine agent i.e., sumatriptin, almotriptan, frovatriptan, sumatriptan, rizatriptan, eletriptan, zolmitriptan, dihydroergotamine, eletriptan and ergotamine
  • acetaminophen a salicylate (i.e., aspirin, choline salicylate, magnesium salicylate, diflunisal, and salsalate), a anti-convulsant (i.e., carbamazepine, clonazepam, gabapentin, lamotrigine, pregabalin, tiagabine, and topiramate), an anaesthetic (i.e., isoflurane, trichloroethylene, halothane, sevoflurane, benzocaine, chloroprocaine, cocaine, cyclomethycaine, dimethocaine, propoxycaine, procaine, novocaine, proparacaine, tetracaine, articaine, bupivacaine, carticaine, cinchocaine, etidocaine, levobupivacaine, lidocaine, mepivacaine, piperocaine, prilocaine, ropivacaine, trimeca
  • neurological drug may be selected that is an anti-vertigo agent including, but not limited to, meclizine, diphenhydramine, promethazine and diazepam.
  • a neurological drug may be selected that is an anti-nausea agent including, but not limited to, promethazine, chlorpromazine, prochlorperazine, trimethobenzamide, and metoclopramide .
  • Amyloidoses are a group of diseases and disorders associated with extracellular proteinaceous deposits in the CNS, including, but not limited to, secondary amyloidosis, age- related amyloidosis, Alzheimer's Disease (AD), mild cognitive impairment (MCI), Lewy body dementia, Down's syndrome, hereditary cerebral hemorrhage with amyloidosis (Dutch type); the Guam Parkinson-Dementia complex, cerebral amyloid angiopathy, Huntington's disease, progressive supranuclear palsy, multiple sclerosis; Creutzfeld Jacob disease, Parkinson's disease, transmissible spongiform encephalopathy, HlV-related dementia, amyotropic lateral sclerosis (ALS), inclusion-body myositis (IBM), and ocular diseases relating to beta-amyloid deposition (i.e., macular degeneration, drusen-related optic neuropathy, and cataract).
  • AD Alzheimer's Disease
  • MCI mild cognitive impairment
  • Lewy body dementia Down
  • a neurological drug may be selected that includes, but is not limited to, an antibody or other binding molecule (including, but not limited to a small molecule, a peptide, an aptamer, or other protein binder) that specifically binds to a target selected from: beta secretase, tau, presenilin, amyloid precursor protein or portions thereof, amyloid beta peptide or oligomers or fibrils thereof, death receptor 6 (DR6), receptor for advanced glycation endproducts (RAGE), parkin, and huntingtin; a cholinesterase inhibitor (i.e., galantamine, donepezil, rivastigmine and tacrine); an NMDA receptor antagonist (i.e., memantine), a monoamine depletor (i.e., tetrabenazine); an ergoloid mesylate; an anticholinergic
  • antiparkinsonism agent i.e., procyclidine, diphenhydramine, trihexylphenidyl, benztropine, biperiden and trihexyphenidyl
  • a dopaminergic antiparkinsonism agent i.e., entacapone, selegiline, pramipexole, bromocriptine, rotigotine, selegiline, ropinirole, rasagiline, apomorphine, carbidopa, levodopa, pergolide, tolcapone and amantadine
  • a tetrabenazine an anti-inflammatory (including, but not limited to, a nonsteroidal anti-inflammatory drug (i.e., indomethicin and other compounds listed above); a hormone (i.e., estrogen, progesterone and leuprolide); a vitamin (i.e., folate and nicotinamide); a dimebolin; a homotaurine (i
  • Cancers of the CNS are characterized by aberrant proliferation of one or more CNS cell (i.e., a neural cell) and include, but are not limited to, glioma, glioblastoma multiforme, meningioma, astrocytoma, acoustic neuroma, chondroma, oligodendroglioma,
  • meduUoblastomas meduUoblastomas, ganglioglioma, Schwannoma, neurofibroma, neuroblastoma, and extradural, intramedullary or intradural tumors.
  • a neurological drug may be selected that is a chemotherapeutic agent.
  • chemotherapeutic agents include alkylating agents such as thiotepa and
  • CYTOXAN® cyclosphosphamide alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa;
  • ethylenimines and methylamelamines including altretamine, triethylenemelamine,
  • trietylenephosphoramide triethiylenethiophosphor-amide and trimethylolomelamine
  • acetogenins especially bullatacin and bullatacinone
  • dronabinol, MARINOL® beta-lapachone
  • lapachol colchicines
  • betulinic acid a camptothecin (including the synthetic analogue topotecan (HYCAMTIN®), CPT-11
  • calicheamicin especially calicheamicin gammall and calicheamicin omegall (see, e.g., Agnew, Chem Intl. Ed. Engl., 33: 183-186 (1994)); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related
  • chromoprotein enediyne antiobiotic chromophores chromoprotein enediyne antiobiotic chromophores
  • aclacinomysins actinomycin
  • elliptinium acetate an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan;
  • lonidainine maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; 2-ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofrran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"- trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine (ELDISINE®, FILDESIN®); dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine;
  • mercaptopurine methotrexate
  • platinum analogs such as cisplatin and carboplatin
  • vinblastine VELBAN®
  • platinum platinum
  • etoposide VP- 16
  • ifosfamide mitoxantrone
  • DMFO difluorometlhylornithine
  • retinoids such as retinoic acid
  • capecitabine XELODA®
  • pharmaceutically acceptable salts, acids or derivatives of any of the above as well as combinations of two or more of the above such as CHOP, an abbreviation for a combined therapy of cyclophosphamide, doxorubicin, vincristine, and prednisolone, and FOLFOX, an abbreviation for a treatment regimen with oxaliplatin (ELOXATF TM) combined with 5-FU and leucovovin.
  • ELOXATF TM oxaliplatin
  • chemotherapeutic agents are anti-hormonal agents that act to regulate, reduce, block, or inhibit the effects of hormones that can promote the growth of cancer, and are often in the form of systemic, or whole-body treatment. They may be hormones themselves. Examples include anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX®
  • tamoxifen EVISTA® raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTON® toremifene; anti-progesterones; estrogen receptor down-regulators (ERDs); agents that function to suppress or shut down the ovaries, for example, leutinizing hormone-releasing hormone (LHRH) agonists such as LUPRON® and ELIGARD® leuprolide acetate, goserelin acetate, buserelin acetate and tripterelin; other anti- androgens such as flutamide, nilutamide and bicalutamide; and aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® megestrol acetate,
  • chemotherapeutic agents includes bisphosphonates such as clodronate (for example, BONEFOS® or OSTAC®), DIDROCAL® etidronate, NE-58095, ZOMETA® zoledronic acid/zoledronate, FOSAMAX® alendronate, AREDIA® pamidronate, SKELID® tiludronate, or ACTONEL® risedronate; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog); antisense oligonucleotides, particularly those that inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, such as, for example, PKC-alpha, Raf, H-Ras, and epidermal growth factor receptor (EGF-R); vaccines such as THERATOPE® vaccine and gene therapy vaccines, for example, ALLOVECTF ® vaccine, LEUVECTIN® vaccine, and VA
  • LURTOTECAN® topoisomerase 1 inhibitor ABARELIX® rmRH
  • lapatinib ditosylate an ErbB-2 and EGFR dual tyrosine kinase small-molecule inhibitor also known as GW572016
  • pharmaceutically acceptable salts, acids or derivatives of any of the above include
  • anti-cancer immunoglobulins including, but not limited to, trastuzumab, pertuzumab, bevacizumab, alemtuxumab, cetuximab, gemtuzumab ozogamicin, ibritumomab tiuxetan, panitumumab and rituximab.
  • antibodies in conjunction with a toxic label or conjugate may be used to target and kill desired cells (i.e.,
  • cancer cells including, but not limited to, tositumomab with a I radiolabel, or trastuzumab emtansine.
  • Ocular diseases or disorders are diseases or disorders of the eye, which for the purposes herein is considered a CNS organ segregated by the BBB.
  • Ocular diseases or disorders include, but are not limited to, disorders of sclera, cornea, iris and ciliary body (i.e., scleritis, keratitis, corneal ulcer, corneal abrasion, snow blindness, arc eye, Thygeson's superficial punctate keratopathy, corneal neovascularisation, Fuchs' dystrophy, keratoconus, keratoconjunctivitis sicca, ulceris and uveitis), disorders of the lens (i.e., cataract), disorders of choroid and retina (i.e., retinal detachment, retinoschisis, hypertensive retinopathy, diabetic retinopathy, retinopathy, retinopathy of prematurity, age-related macular degeneration, macular
  • degeneration (wet or dry), epiretinal membrane, retinitis pigmentosa and macular edema), glaucoma, floaters, disorders of optic nerve and visual pathways (i.e., Leber's hereditary optic neuropathy and optic disc drusen), disorders of ocular muscles/binocular movement accommodation/refraction (i.e., strabismus, ophthalmoparesis, progressive external opthalmoplegia, esotropia, exotropia, hypermetropia, myopia, astigmatism, anisometropia, presbyopia and ophthalmoplegia), visual disturbances and blindness (i.e., amblyopia, Lever's congenital amaurosis, scotoma, color blindness, achromatopsia, nyctalopia, blindness, river blindness and micro-opthalmia/coloboma), red eye, Argyll Robertson pupil, keratomy
  • a neurological drug may be selected that is an anti- angiogenic ophthalmic agent (i.e., bevacizumab, ranibizumab and pegaptanib), an ophthalmic glaucoma agent (i.e., carbachol, epinephrine, demecarium bromide, apraclonidine, brimonidine, brinzolamide, levobunolol, timolol, betaxolol, dorzolamide, bimatoprost, carteolol, metipranolol, dipivefrin, travoprost and latanoprost), a carbonic anhydrase inhibitor (i.e., methazolamide and acetazolamide), an ophthalmic antihistamine (i.e., naphazoline, phenylephrine and tetrahydrozoline), an ocular lubricant, an ophthalmic agent (i.e.,
  • Viral or microbial infections of the CNS include, but are not limited to, infections by viruses (i.e., influenza, HIV, poliovirus, rubella, ), bacteria (i.e., Neisseria sp., Streptococcus sp., Pseudomonas sp., Proteus sp., E. coli, S.
  • viruses i.e., influenza, HIV, poliovirus, rubella,
  • bacteria i.e., Neisseria sp., Streptococcus sp., Pseudomonas sp., Proteus sp., E. coli, S.
  • aureus Pneumococcus sp., Meningococcus sp., Haemophilus sp., and Mycobacterium tuberculosis
  • fungi i.e., yeast, Cryptococcus neoformans
  • parasites i.e., toxoplasma gondii
  • amoebas resulting in CNS pathophysiologies including, but not limited to, meningitis, encephalitis, myelitis, vasculitis and abscess, which can be acute or chronic.
  • a neurological drug may be selected that includes, but is not limited to, an antiviral compound (including, but not limited to, an adamantane antiviral (i.e., rimantadine and amantadine), an antiviral interferon (i.e., peginterferon alfa-2b), a chemokine receptor antagonist (i.e., maraviroc), an integrase strand transfer inhibitor (i.e., raltegravir), a neuraminidase inhibitor (i.e., oseltamivir and zanamivir), a non-nucleoside reverse transcriptase inhibitor (i.e., efavirenz, etravirine, delavirdine and nevirapine), a nucleoside reverse transcriptase inhibitors (tenofovir, abacavir, lamivudine, zidovudine, stavudine, ente
  • an antiviral compound including,
  • vancomycin vancomycin
  • a macrolide i.e., azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin and spectinomycin
  • a monobactam i.e., aztreonam
  • a quinolone i.e., ciprofloxacin, enoxacin, gatifloxacin, levofioxacin
  • lomefloxacin moxifloxacin, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin and temafloxacin
  • a sulfonamide i.e., mafenide, sulfonamidochrysoidine, sulfacetamide, sulfadiazine, sulfamethizole, sulfanilamide, sulfasalazine, sulfisoxazole, trimethoprim, trimethoprim and sulfamethoxazole
  • a tetracycline i.e., tetracycline, demeclocycline, doxycycline, minocycline and oxytetracycline
  • an antineoplastic or cytotoxic antibiotic i.e., doxorubicin, mitoxantrone, bleomycin, daunorubicin, d
  • Inflammation of the CNS includes, but is not limited to, inflammation that is caused by an injury to the CNS, which can be a physical injury (i.e., due to accident, surgery, brain trauma, spinal cord injury, concussion) and an injury due to or related to one or more other diseases or disorders of the CNS (i.e., abscess, cancer, viral or microbial infection).
  • an injury to the CNS which can be a physical injury (i.e., due to accident, surgery, brain trauma, spinal cord injury, concussion) and an injury due to or related to one or more other diseases or disorders of the CNS (i.e., abscess, cancer, viral or microbial infection).
  • a neurological drug may be selected that addresses the inflammation itself (i.e., a nonsteroidal anti-inflammatory agent such as ibuprofen or naproxen), or one which treats the underlying cause of the inflammation (i.e., an anti-viral or anti-cancer agent).
  • a nonsteroidal anti-inflammatory agent such as ibuprofen or naproxen
  • an anti-viral or anti-cancer agent i.e., an anti-viral or anti-cancer agent
  • Ischemia of the CNS refers to a group of disorders relating to aberrant blood flow or vascular behavior in the brain or the causes therefor, and includes, but is not limited to: focal brain ischemia, global brain ischemia, stroke (i.e., subarachnoid hemorrhage and intracerebral hemorrhage), and aneurysm.
  • a neurological drug may be selected that includes, but is not limited to, a thrombolytic (i.e., urokinase,reteplase, reteplase and tenecteplase), a platelet aggregation inhibitor (i.e., aspirin, cilostazol, clopidogrel, prasugrel and dipyridamole), a statin (i.e., lovastatin, pravastatin, fluvastatin, rosuvastatin, atorvastatin, simvastatin, cerivastatin and pitavastatin), and a compound to improve blood flow or vascular flexibility, including, e.g., blood pressure medications.
  • a thrombolytic i.e., urokinase,reteplase, reteplase and tenecteplase
  • a platelet aggregation inhibitor i.e., aspirin, cilostazol,
  • Neurodegenerative diseases are a group of diseases and disorders associated with neural cell loss of function or death in the CNS, and include, but are not limited to:
  • adrenoleukodystrophy Alexander's disease, Alper's disease, amyotrophic lateral sclerosis, ataxia telangiectasia, Batten disease, cockayne syndrome, corticobasal degeneration, degeneration caused by or associated with an amyloidosis, Friedreich's ataxia, frontotemporal lobar degeneration, Kennedy's disease, multiple system atrophy, multiple sclerosis, primary lateral sclerosis, progressive supranuclear palsy, spinal muscular atrophy, transverse myelitis, Refsum's disease, and spinocerebellar ataxia.
  • a neurological drug may be selected that is a growth hormone or neurotrophic factor; examples include but are not limited to brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-4/5, fibroblast growth factor (FGF)-2 and other FGFs, neurotrophin (NT)-3, erythropoietin (EPO), hepatocyte growth factor (HGF), epidermal growth factor (EGF), transforming growth factor (TGF)-alpha, TGF- beta, vascular endothelial growth factor (VEGF), interleukin-1 receptor antagonist (IL-lra), ciliary neurotrophic factor (CNTF), glial-derived neurotrophic factor (GDNF), neurturin, platelet-derived growth factor (PDGF), heregulin, neuregulin, artemin, persephin, interleukins, glial cell line derived neurotrophic factor (GFR), granulocyte-colony stimulating factor (CSF), granulocyte-
  • BDNF brain-
  • Seizure diseases and disorders of the CNS involve inappropriate and/or abnormal electrical conduction in the CNS, and include, but are not limited to epilepsy (i.e., absence seizures, atonic seizures, benign Rolandic epilepsy, childhood absence, clonic seizures, complex partial seizures, frontal lobe epilepsy, febrile seizures, infantile spasms, juvenile myoclonic epilepsy, juvenile absence epilepsy, Lennox-Gastaut syndrome, Landau- leffner Syndrome, Dravet's syndrome, Otahara syndrome, West syndrome, myoclonic seizures, mitochondrial disorders, progressive myoclonic epilepsies, psychogenic seizures, reflex epilepsy, Rasmussen's Syndrome, simple partial seizures, secondarily generalized seizures, temporal lobe epilepsy, toniclonic seizures, tonic seizures, psychomotor seizures, limbic epilepsy, partial-onset seizures, generalized-onset seizures, status epilepticus, abdominal epilepsy, akinetic seizures, autonomic seizures, massive bilateral myoclo
  • a neurological drug may be selected that is an anticonvulsant or antiepileptic including, but not limited to, barbiturate anticonvulsants (i.e., primidone, metharbital, mephobarbital, allobarbital, amobarbital, aprobarbital, alphenal, barbital, brallobarbital and phenobarbital), benzodiazepine anticonvulsants (i.e., diazepam, clonazepam, and lorazepam), carbamate anticonvulsants (i.e. felbamate), carbonic anhydrase inhibitor anticonvulsants (i.e., acetazolamide, topiramate and zonisamide), dibenzazepine
  • barbiturate anticonvulsants i.e., primidone, metharbital, mephobarbital, allobarbital, amobarbital, aprobarbital, alphenal, barbital
  • anticonvulsants i.e., rafmamide, carbamazepine, and oxcarbazepine
  • fatty acid derivative anticonvulsants i.e., divalproex and valproic acid
  • gamma-aminobutyric acid analogs i.e., pregabalin, gabapentin and vigabatrin
  • gamma-aminobutyric acid reuptake inhibitors i.e., tiagabine
  • gamma-aminobutyric acid transaminase inhibitors i.e., vigabatrin
  • hydantoin anticonvulsants i.e.
  • phenytoin, ethotoin, fosphenytoin and mephenytoin miscellaneous anticonvulsants (i.e., lacosamide and magnesium sulfate), progestins (i.e., progesterone), oxazolidinedione anticonvulsants (i.e., paramethadione and trimethadione), pyrrolidine anticonvulsants (i.e., levetiracetam), succinimide anticonvulsants (i.e., ethosuximide and methsuximide), triazine anticonvulsants (i.e., lamotrigine), and urea anticonvulsants (i.e., phenacemide and pheneturide).
  • progestins i.e., progesterone
  • oxazolidinedione anticonvulsants i.e., paramethadione and trimethadione
  • Behavioral disorders are disorders of the CNS characterized by aberrant behavior on the part of the afflicted subject and include, but are not limited to: sleep disorders (i.e., insomnia, parasomnias, night terrors, circadian rhythm sleep disorders, and narcolepsy), mood disorders (i.e., depression, suicidal depression, anxiety, chronic affective disorders, phobias, panic attacks, obsessive-compulsive disorder, attention deficit hyperactivity disorder (ADHD), attention deficit disorder (ADD), chronic fatigue syndrome, agoraphobia, post-traumatic stress disorder, bipolar disorder), eating disorders (i.e., anorexia or bulimia), psychoses,
  • sleep disorders i.e., insomnia, parasomnias, night terrors, circadian rhythm sleep disorders, and narcolepsy
  • mood disorders i.e., depression, suicidal depression, anxiety, chronic affective disorders, phobias, panic attacks, obsessive-compulsive disorder, attention deficit hyperactivity disorder (ADHD), attention
  • developmental behavioral disorders i.e., autism, Rett's syndrome, Aspberger's syndrome
  • personality disorders i.e., schizophrenia, delusional disorder, and the like.
  • a neurological drug may be selected from a behavior- modifying compound including, but not limited to, an atypical antipsychotic (i.e., risperidone, olanzapine, apripiprazole, quetiapine, paliperidone, asenapine, clozapine, iloperidone and ziprasidone), a phenothiazine antipsychotic (i.e., prochlorperazine, chlorpromazine, fluphenazine, perphenazine, trifluoperazine, thioridazine and mesoridazine), a thioxanthene (i.e., thiothixene), a miscellaneous antipsychotic (i.e., pimozide, lithium, molindone, haloperidol and loxapine), a selective serotonin reuptake inhibitor (i.e., citalopram, escitalopram, es
  • phenylpiperazine antidepressant i.e., trazodone and nefazodone
  • a monoamine oxidase inhibitor i.e., isocarboxazid, phenelzine, selegiline and tranylcypromine
  • a benzodiazepine i.e., alprazolam, estazolam, flurazeptam, clonazepam, lorazepam and diazepam
  • a benzodiazepine i.e., alprazolam, estazolam, flurazeptam, clonazepam, lorazepam and diazepam
  • norepinephrine-dopamine reuptake inhibitor i.e., bupropion
  • a CNS stimulant i.e., phentermine, diethylpropion, methamphetamine, dextroamphetamine, amphetamine, methylphenidate, dexmethylphenidate, lisdexamfetamine, modafinil, pemoline,
  • phendimetrazine benzphetamine, phendimetrazine, armodafmil, diethylpropion, caffeine, atomoxetine, doxapram, and mazindol
  • anxiolytic/sedative/hypnotic including, but not limited to, a barbiturate (i.e., secobarbital, phenobarbital and mephobarbital), a benzodiazepine (as described above), and a miscellaneous anxiolytic/sedative/hypnotic (i.e.
  • Lysosomal storage disorders are metabolic disorders which are in some cases associated with the CNS or have CNS-specific symptoms; such disorders include, but are not limited to: Tay-Sachs disease, Gaucher' s disease, Fabry disease, mucopolysaccharidosis (types I, II, III, IV, V, VI and VII), glycogen storage disease, GM1 -gangliosidosis, metachromatic
  • lipofuscinoses types 1 and 2 Niemann-Pick disease, Pompe disease, and Krabbe's disease.
  • a neurological drug may be selected that is itself or otherwise mimics the activity of the enzyme that is impaired in the disease.
  • Exemplary recombinant enzymes for the treatment of lysosomal storage disorders include, but are not limited to those set forth in e.g., U.S. Patent Application publication no.
  • 2005/0142141 i.e., alpha-L-iduronidase, iduronate-2-sulphatase, N-sulfatase, alpha-N-acetylglucosaminidase, N- acetyl-galactosamine-6-sulfatase, beta-galactosidase, arylsulphatase B, beta-glucuronidase, acid alpha-glucosidase, glucocerebrosidase, alpha-galactosidase A, hexosaminidase A, acid sphingomyelinase, beta-galactocerebrosidase, beta-galactosidase, arylsulfatase A, acid ceramidase, aspartoacylase, palmitoyl-protein thioesterase 1 and tripeptidyl amino peptidase 1).
  • diseases related to or caused by inappropriate overproduction of red blood cells, or wherein the overproduction of red blood cells is an effect of the disease can be prevented or treated by the reticulocyte-depleting effect recognized herein of anti-Tf antibodies retaining at least partial effector function.
  • the reticulocyte-depleting effect recognized herein of anti-Tf antibodies retaining at least partial effector function.
  • elevated red blood cell counts due to hyperproliferation of, e.g., reticulocytes results in thickening of blood and concomitant physiological symptoms (d'Onofrio et al., Clin. Lab. Haematol. (1996) Suppl. 1 : 29-34).
  • an anti-TfR antibody of the invention wherein at least partial effector function of the antibody was preserved would permit selective removal of immature reticulocyte populations without impacting normal transferrin transport into the CNS. Dosing of such an antibody could be modulated such that acute clinical symptoms could be minimized (ie, by dosing at a very low dose or at widely-spaced intervals), as well-understood in the art.
  • an antibody of the invention is used to detect a neurological disorder before the onset of symptoms and/or to assess the severity or duration of the disease or disorder.
  • the antibody permits detection and/or imaging of the neurological disorder, including imaging by radiography, tomography, or magnetic resonance imaging (MRI).
  • MRI magnetic resonance imaging
  • a low affinity anti-TfR antibody of the invention for use as a medicament is provided.
  • a low affinity anti-TfR antibody for use in treating a neurological disease or disorder e.g., Alzheimer's disease
  • red blood cells ie, reticulocytes
  • a modified low affinity anti-TfR antibody for use in a method of treatment as described herein is provided.
  • the invention provides a low affinity anti-TfR antibody modified to improve its safety for use in a method of treating an individual having a neurological disease or disorder comprising administering to the individual an effective amount of the anti-TfR antibody (optionally coupled to a neurological disorder drug). In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent. In further embodiments, the invention provides an anti-TfR antibody modified to improve its safety for use in reducing or inhibiting amlyoid plaque formation in a patient at risk or suffering from a neurological disease or disorder (e.g., Alzheimer's disease). An "individual" according to any of the above embodiments is optionally a human. In certain aspects, the anti-TfR antibody of the invention for use in the methods of the invention improves uptake of the neurological disorder drug with which it is coupled.
  • the invention provides for the use of a low affinity anti-TfR antibody of the invention in the manufacture or preparation of a medicament.
  • the medicament is for treatment of neurological disease or disorder.
  • the medicament is for use in a method of treating neurological disease or disorder comprising administering to an individual having neurological disease or disorder an effective amount of the medicament. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent.
  • the invention provides a method for treating Alzheimer's disease.
  • the method comprises administering to an individual having Alzheimer's disease an effective amount of a multispecific antibody of the invention which binds both BACE1 and TfR or both Abeta and TfR.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent.
  • An "individual" according to any of the above embodiments may be a human.
  • the anti-TfR antibodies of the invention can be used either alone or in combination with other agents in a therapy.
  • the anti-TfR antibody of the invention may be coadministered with at least one additional therapeutic agent.
  • an additional therapeutic agent is a therapeutic agent effective to treat the same or a different neurological disorder as the anti-TfR antibody is being employed to treat.
  • Exemplary additional therapeutic agents include, but are not limited to: the various neurological drugs described above, cholinesterase inhibitors (such as donepezil, galantamine, rovastigmine, and tacrine), NMDA receptor antagonists (such as memantine), amyloid beta peptide aggregation inhibitors, antioxidants, ⁇ -secretase modulators, nerve growth factor (NGF) mimics or NGF gene therapy, PPARy agonists, HMS-CoA reductase inhibitors (statins), ampakines, calcium channel blockers, GABA receptor antagonists, glycogen synthase kinase inhibitors, intravenous immunoglobulin, muscarinic receptor agonists, nicrotinic receptor modulators, active or passive amyloid beta peptide immunization, phosphodiesterase inhibitors, serotonin receptor antagonists and anti-amyloid beta peptide antibodies.
  • the at least one additional therapeutic agent is selected for its ability to mitigate one or more side effects of the neurological drugs described
  • certain anti-TfR antibodies may have side effects that negatively impact reticulocyte populations in a subject treated with the anti-TfR antibody.
  • at least one further therapeutic agent selected for its ability to mitigate such negative side effect on reticulocyte populations is coadministered with an anti-TfR antibody of the invention.
  • therapeutic agents include, but are not limited to, agents to increase red blood cell (ie, reticulocyte) populations, agents to support growth and development of red blood cells (ie, reticulocytes), and agents to protect red blood cell populations from the effects of the anti-TfR antibody; such agents include, but are not limited to, erythropoietin (EPO), iron supplements, vitamin C, folic acid, and vitamin B12, as well as physical replacement of red blood cells (ie, reticulocytes) by, for example, transfusion with similar cells, which may be from another individual of similar blood type or may have been previously extracted from the subject to whom the anti-TfR antibody is administered.
  • EPO erythropoietin
  • iron supplements iron supplements
  • vitamin C folic acid
  • vitamin B12 vitamin B12
  • agents intended to protect existing red blood cells are preferably administered to the subject preceding or concurrent with the anti-TfR antibody therapy, while agents intended to support or initiate the regrowth/development of red blood cells or blood cell populations (ie, reticulocytes or reticulocyte populations) are preferably administered concurrent with or after the anti-TfR antibody therapy such that such blood cells can be replenished after the anti-TfR antibody treatment.
  • the at least one further therapeutic agent is selected for its ability to inhibit or prevent the activation of the complement pathway upon
  • anti-TfR antibody examples include, but are not limited to, agents that interfere with the ability of the anti-TfR antibody to bind to or activate the complement pathway and agents that inhibit one or more molecular interactions within the complement pathway, and are described generally in Mollnes and Kirschftnk (2006) Molec. Immunol. 43:107-121, the contents of which are expressly incorporated herein by reference.
  • administration where two or more therapeutic agents are included in the same or separate formulations
  • separate administration in which case, administration of the antibody of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
  • administration of the anti-TfR antibody and administration of an additional therapeutic agent occur within about one month, or within about one, two or three weeks, or within about one, two, three, four, five or six days, of each other.
  • Antibodies of the invention can also be used in combination with other interventional therapies such as, but not limited to, radiation therapy, behavioral therapy, or other therapies known in the art and appropriate for the neurological disorder to be treated or prevented.
  • An anti-TfR antibody of the invention can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • Various dosing schedules including but not limited to single or multiple administrations over various time- points, bolus administration, and pulse infusion are contemplated herein.
  • Antibodies of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question or to prevent, mitigate or ameliorate one or more side effects of antibody administration.
  • the effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.
  • an antibody of the invention when used alone or in combination with one or more other additional therapeutic agents, will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the antibody is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to 15 mg/kg (e.g. O. lmg/kg-lOmg/kg) of antibody can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • One typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs.
  • One exemplary dosage of the antibody would be in the range from about 0.05 mg/kg to about 40 mg/kg.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg, 5.0 mg/kg, 7.5 mg/kg, 10 mg/kg, 15 mg/kg, 20 mg/kg, 25 mg/kg, 30 mg/kg, 35 mg/kg or 40 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermittently, e.g.
  • a lower dose of the anti-TfR antibodies may be administered with greater frequency than a higher dose would be.
  • the dosage used may be balanced between the amount of antibody necessary to be delivered to the CNS (itself related to the affinity of the CNS antigen-specific portion of the antibody), the affinity of that antibody for TfR, and whether or not red blood cell (ie, reticulocyte)-protecting, growth and
  • development-stimulating, or complement pathway-inhibiting compound(s) are being co- or serially administered with the antibody.
  • the progress of this therapy is easily monitored by conventional techniques and assays as described herein and as known in the art. It is understood that any of the above formulations or therapeutic methods may be carried out using an immunoconjugate of the invention in place of or in addition to an anti-Tf antibody.
  • an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is an antibody of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an antibody of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bac
  • any of the above articles of manufacture may include an immunoconjugate of the invention in place of or in addition to an anti-TfR antibody.
  • EXAMPLE 1 GENERATION, CHARACTERIZATION AND HUMANIZATION OF HUMAN/CYNO CROSS-REACTIVE ANTI-TFR ANTIBODIES
  • a na ' ive antibody phage panning process was performed in an attempt to identify antibodies cross-reactive with both human TfR and TfR from cynomolgous ("cyno") monkeys that further did not compete with Tf for binding to TfR (Lee et al. JMB (2004) 1073- 1093). No such cross-reactive, non-Tf-competing clone was identified from this phage panning process. However, two antibodies were identified that were useful in characterizing subsequently generated hybridoma clones.
  • Tf-competing antibody A species cross-reactive antibody was identified that competes with Tf for binding to human or cyno TfR (Tf-competing antibody).
  • Tf-competing antibody The epitope of another clone, specific for human TfR, was mapped to the apical domain of huTfR using mouse/human chimeric TfR receptors ( Figure 1). This apical domain-binding clone lost binding to huTfR when the mouse TfR sequence in the apical domain was substituted into huTfR.
  • Human and cyno cross-reactive TfR antibodies were generated by immunizing 5 Balb/C mice in the footpad with 6 doses (twice per week) containing 2 ⁇ g each of cynoTfR and huTfR ecd. All mice sera were FACS positive and all mice were fused. Of 1632 hybridomas screened, 1 1 1 were ELISA positive for binding to both human and cyno TfR.
  • the resulting ELISA-positive hybridomas were screened by FACS in the presence of 1 ⁇ human holo-Tf for binding to 293 cells transiently expressing human or cyno TfR. Briefly, FACS analysis was performed using 293 cells transfected with full length human or cyno TfR using lipofectamin 2000 plus (Invitrogen) 48-72 h before FACS analysis.
  • Non-transfected (control) and transfected 293 cells were washed twice with FACS buffer (PBS containing 1% BSA), 50 ⁇ of hybridoma supernatant (normalized to 10 ⁇ g/ml) was added to 293 cells in the presence of 1 ⁇ human holo-Tf and incubated on ice for 30 min. Cells were washed twice with FACS buffer, 50 ⁇ of PE-Goat-anti-murine Fey (Jackson ImmunoResearch) was added to cells and they were incubated on ice for 30 min. Cells were washed with FACS buffer and resuspended in ⁇ FACS buffer for analysis.
  • FACS buffer PBS containing 1% BSA
  • Antibody affinities were measured using surface plasmon resonance (“SPR”)
  • Anti-His antibody (Qiagen) was coupled onto four different flow cells of a BIACORETM CM5 sensor chip (Biacore, Inc., Piscataway, NJ) at between 6000 and 8000 RU. Immobilization was achieved by random coupling through amino groups using a protocol provided by the manufacturer. 10X HBS-P (Biacore, Inc., Piscataway, NJ) was diluted in water and served as the dilution and running buffer. Purified human or cyno TfR was captured, followed by a 3 -fold dilution series of IgG or Fab that was injected at a flow rate of 30 ml/min using the single cycle kinetics method.
  • Affinity constants were determined using a simple 1 : 1 Langmuir binding model or using a steady state model when k on or k 0 ff was beyond the detection limit.
  • the equilibrium dissociation constant (K D ) was calculated as the ratio of association rate constant (k on ) and dissociation rate constant (k 0 ff). The results are shown in Figure 2C.
  • 7G7 was humanized by grafting the HVRs into the kappa 4 and subgroup I human consensus variable domains along with selected vernier positions in VH (position 93) as outlined in Figure 4C.
  • This humanized variant is called 7G7.vl.
  • 16F6 was humanized by grafting the HVRs into the IGKV1-9*01 and IGHV4-59*01 human variable domains. Combinations of different mouse vernier positions were included in the humanized variants as outlined in Figure 4E .
  • Humanized 16F6 variant 16F6.v4 contains 2 changes in VL (I48L and F71Y) as well as selected vernier positions in VL (positions 43 and 44) and VH (positions 71 and 78) as outlined in Figure 4D.
  • the binding epitope of the antibodies were re-confirmed as follows.
  • a Tf-TfR blocking ELISA was performed in maxisorp plates coated with 2 ⁇ of purified human TfR in PBS at 4°C overnight. Plates were washed with PBS/0.05% Tween 20 and blocked using Superblock blocking buffer in PBS (Thermo Scientific, Hudson, NH). 50 ⁇ of 12.5 ⁇ human holo-Tf (R&D Systems, Minneapolis, MN) was added to the plates for 40 min.
  • HFE-TfR binding ELISA was performed in maxisorp plates coated with 1 ⁇ of HFE in PBS at 4°C overnight. Plates were washed with PBS/0.05% Tween 20 and blocked using Superblock blocking buffer in PBS (Thermo Scientific, Hudson, NH). A titration of human TfR (start at 100 ug ml, 1 :3 serial dilution) was added to the plate and incubated for 1 h. ⁇ g/ml of hul Gl l.v5, hu7A4.vl5 or hu7G7.vl was then added to the plate for 1 h.
  • a titration of hu7A4.vl5, hul5Gl l .v5, Tf competiting antibody, human holo- Tf and control IgG 400 ⁇ g for all antibody, 8000 ⁇ g/ml for holo transferrin, 1 :3 serial dilution
  • hu7A4.vl5, hul5Gl l .v5, Tf competiting antibody, human holo- Tf and control IgG 400 ⁇ g for all antibody, 8000 ⁇ g/ml for holo transferrin, 1 :3 serial dilution
  • the mixture was then added to the HFE coated plate for 1 h at room temperature.
  • Immobilized 15Gl lv.5 and anti-TfR were evaluated for binding to biotinylated human TfR ECD or monovalent M13 phage displaying the human TfR apical domain.
  • Anti- TfR 012 was derived from a synthetic antibody phage library that was panned against human TfR ECD and binds to a site on the human TfR which competes with transferrin binding.
  • Antibodies were coated at 1 ⁇ g/ml in PBS on Maxisorp plates.
  • Bound biotinylated human TfR ECD or TfR-apical domain phage were detected with HRP-streptavidin (GE health care, RPN 4401V) or HRP-anti-M13 (GE health care, 27-9421-01), respectively.
  • Figure 25 shows that 15Gl lv.5 binds to human TfR apical domain. The 15Gl lv.5 binding site was mapped to the apical domain, a site distant from the TfR ligand binding sites.
  • EXAMPLE 2 AFFINITY ENGINEERING HUMAN/ CYNO CROSS-REACTIVE ANTI- TFR ANTIBODIES
  • affinity engineered variants were made. Exemplified herein is affinity engineering of 15Gl l .v5 and 7A4.vl5. Affinity variants were generated by making individual alanine substitutions in CDR-L3 or CDR-H3 using standard techniques. These variants were screened as IgG by ELISA and SPR to identify positions important for binding to human and cyno TfR; the monovalent affinity of selected variants as Fab was also determined.
  • Cell culture supernatants containing the expressed Ala scan variant IgGs were serially diluted 1 :5 and added to the wells for 1 h. Plates were washed with PBS/0.05% Tween 20 and 1 : 1000 diluted HRP-Goat-anti human Fey (Jackson ImmunoResearch) was added the plate and incubated for 1 hour at room temperature. Plates were washed with PBS/0.05% Tween 20 and detected using TMB substrate (BioFX Laboratories, Owings Mills).
  • Certain of the foregoing antibody variants were reformatted as bispecific antibodies with a second arm specifically binding to BACE1.
  • the anti-human TfR antibodies Hul5Gl l .v5, Hul5Gl l .LC92A , Hul5Gl l .HC52A and Hul5Gl 1.HC53A were used to engineer the TfR binding arm of the bispecific using 'knob in hole' bispecific antibody construction technology (Carter, P. (2001) J. Immunol. Methods 248, 7-15; Ridgway, J. B., Presta, L. G., and Carter, P. (1996) Protein Eng. 9, 617-621 ; Merchant, A. M., Zhu, Z., Yuan, J.
  • the knob and hole half-antibodies were purified separately from E.coli and combined at a 1 : 1.1 ratio of anti-TfR to prevent formation of anti-TfR homodimers. Assembly of the bispecific antibody was completed by reductive annealing for at least three days at room temperature in a buffer containing reduced glutathione at a lOOx ratio to antibody and 200 mM arginine pH 8.0. Following assembly, bispecific antibodies were purified by hydrophobic interaction chromatography. The assembly was confirmed by liquid chromatography mass spectroscopy and SDS-PAGE. The purified antibodies were confirmed to be homogeneous and monodisperse by size exclusion and multi angle laser light spectroscopy.
  • the resulting bispecific antibodies were called 15Gl l.v5 (anti-TfR 1 ), 15G11.W92A (15G11.LC92A or anti-TfR 2 ), Hul5Gl l.N52A (anti-TfR 52A ) and Hul5Gl l .T53A (anti- TfR 53A ).
  • the monovalent affinity and kinetics for human and cyno TfR was determined for 115G11.V5 and 115G11.W92A by SPR, as above (see Table 9).
  • Anti-TfR 1 and anti-TfR 2 possess similar monovalent binding affinities as anti-TfR A and anti-TfR D do for binding to mouse TfR (see Atwal et al, Sci. Transl. Med. 3, 84ra43 (2011); Yu et al, Sci. Transl. Med. 25 May 2011 : Vol. 3, Issue 84, p. 84ra44).
  • the binding affinity of the anti-TfR 1 , anti-TfR 2 , Hul5Gl l .N52A and Hul5Gl l .T53A bispecific antibodies were measured against human and cyno TfR by SPR as previously described. As shown in Table 9 below, Anti-TfR 52A and anti-TfR 53A have binding affinities to human and cyno TfR between TfRl hl5Gl l v5 and TfR2 LC9 A .
  • EXAMPLE 4 IMPACT OF EFFECTOR-CONTAINING AND EFFECTORLESS MONOSPECIFIC AND BISPECIFIC ANTIBODIES ON A HUMAN ERYTHROLEUKEMIA CELL LINE AND PRIMARY BONE MARROW MONONUCLEAR CELLS
  • mice Prior studies in mice had determined that antibodies binding murine TfR with effector function and/or complement binding capabilities selectively depleted TfR-expressing reticulocytes. To ascertain whether the depletion observed in the mouse studies was unique to a murine system, further experiments were performed utilizing anti-TfR that bind to human TfR.
  • ADCC assays were carried out using peripheral blood mononuclear cells (PBMCs) from healthy human donors as effector cells.
  • PBMCs peripheral blood mononuclear cells
  • HEL human erythroleukemia cell line
  • AllCells, Inc. primary human bone marrow mononuclear cells
  • PBMCs were isolated by density gradient centrifugation using Uni-SepTM blood separation tubes (Accurate Chemical & Scientific Corp.; Westbury, NY).
  • Target cells in 50 ⁇ , of assay medium (RPMI-1640 with 1% BSA and 100 units/mL penicillin and streptomycin) were seeded in a 96-well, round-bottom plate at 4 x 10 4 /well.
  • Serial dilutions of test and control antibodies 50 ⁇ were added to the plates containing the target cells, followed by incubation at 37 C with 5% C0 2 for 30 minutes to allow opsonization.
  • the final concentrations of antibodies ranged from 0.0051 to 10,000 ng/mL following 5-fold serial dilutions for a total of 10 data points.
  • ADCC values of sample dilutions were plotted against the antibody concentration, and the dose-response curves were fitted to a four-parameter model using SoftMax Pro.
  • the ADCC activity of various anti-human TfR constructs were assessed using either a human erythroleukemia cell line (HEL cells) or primary human bone marrow mononuclear cells as the target cells.
  • HEL cells human erythroleukemia cell line
  • the somewhat lower level observed in the bone marrow mononuclear cells experiment is likely due to the fact that only a portion of the heterogeneous mixture of myeloid and erythroid lineage PBMC cells used in the experiment express high levels of TfR, whereas the HEL cells have consistently high TfR expression throughout the clonal cell population.
  • the bispecific effectorless anti-humanTfR/BACEl antibody did not display any ADCC activity in either HEL or bone marrow mononuclear cells, similar to the negative control.
  • Anti-TfR/gD with the wild-type IgGl induced robust ADCC in HEL cells, while the anti-TfR gD with the effectorless IgGl did not show any ADCC activity in HEL cells, replicating the results from the first set of experiments.
  • anti-TfR/gD of the IgG4 isotype showed a mild ADCC activity. This activity was not observed in the anti- Abeta IgG4 results, indicating that TfR binding was required for the ADCC activity. This finding correlates with previous reports that IgG4 has minimal, but measurable, effector function (Adolffson et al, J. Neurosci.
  • cynomolgus monkeys (Macaca fascicular is) were dosed with bispecific antibodies using anti-TfR antibody clone 15G11 paired with the same anti-BACEl arm used in prior examples (anti-TfRVBACEl), or clone 15G11.LC92A paired with the same anti-BACEl arm used in prior examples (anti-TfR 2 /BACEl) or Hul5Gl l .N52A (anti-TfR 52A /BACEl) and Hul5Gl l .T 53A (anti-TfR 53A /BACEl).
  • bispecific antibodies were in a human IgGl format with N297G or D265A and N297G mutations abrogating effector function, as described previously.
  • an anti-gD molecule on human IgGl was used. This study was performed in non-human primates because crossreactivity of these anti-TfR antibodies is limited to non-human primates and humans.
  • studies have shown that the mechanisms of drug transport between the cerebrospinal fluid (CSF) and plasma compartments may be similar between humans and primates (Poplack et al, 1977).
  • CSF cerebrospinal fluid
  • the antibodies were administered by a single intravenous (IV) bolus injection into the saphenous vein at a dose of 30 mg/kg to conscious cynomolgus monkeys with indwelling cisterna magna catheters.
  • IV intravenous
  • CSF plasma, serum, and
  • the concentrations of the dosed antibodies in cynomolgus monkey serum and CSF were measured with an ELISA using a sheep anti-human IgG monkey absorbed antibody coat, followed by adding serum samples starting at a dilution of 1 : 100, and finished by adding a goat anti-human IgG antibody conjugated to horseradish peroxidase monkey adsorbed for detection.
  • the assay had a standard curve range of 0.78-50 ng/mL and a limit of detection of 0.08 ⁇ g/mL. Results below this limit of detection were reported as less than reportable (LTR).
  • Figures 11A-B shows the results of the pharmacokinetic analysis for anti-TfRl/BACEl and anti-TfR2/BACEl .
  • the pharmacokinetic profile for anti-gD was as expected for a typical human IgGl antibody in cynomolgus monkey with a mean clearance of 3.98 mL/day/kg. Both anti-TfR/BACEl antibodies cleared faster than anti-gD, likely due to peripheral target- mediated clearance.
  • Anti-TfRl/BACEl had the fastest clearance, consistent with it having the highest binding affinity to TfR, whereas anti-TfR2/BACEl showed an improved pharmacokinetic profile (ie, prolonged exposure in serum) as compared to anti-TfRl/BACEl, likely due to its reduced affinity for TfR.
  • the clearance for anti-TfRl/BACEl and anti- TfR2/BACEl were 18.9 mL/day/kg and 8.14 mL/day/kg, respectively. All antibodies were detected in the CSF at approximately one one-thousandth of the serum concentration. However, there was high variability, and overall no detectable difference in the CSF antibody concentrations across the molecules.
  • SD standard deviation
  • IV intravenous
  • AUC a n area under the concentration-time curve from time 0 to the time of last measurable concentration
  • AUQ nf area under the concentration-time curve extrapolated to infinity
  • C max observed maximum serum concentration
  • CL clearance
  • V ss volume of distribution at steady state
  • Min minimum
  • Max maximum.
  • Figure 19 shows the results of the pharmacokinetic analysis for anti-TfRVBACEl, anti- TfR 52A /BACEl and anti-TfR 53A /BACE 1. All anti-TfR/BACEl antibodies cleared faster than anti-gD, likely due to peripheral target-mediated clearance. Anti-TfRVBACEl had the fastest clearance, consistent with it having the highest binding affinity to TfR, whereas anti- TfR 52A /BACEl and anti-TfR 5 A /BACEl showed an improved pharmacokinetic profile (ie, prolonged exposure in serum) as compared to anti-TfRVBACEl, likely due to the reduced affinity for TfR of anti-TfR 52A /BACEl and anti-TfR 5 A /BACEl .
  • Abetai_ 40 and sAPPa and ⁇ levels in cynomolgus monkey plasma and CSF were measured.
  • Abetai_4o was measured with an ELISA using an anti-Abetai_4o specific polyclonal antibody coat, followed by adding samples, and finishing by adding a mouse anti-human Abetai_ 40 monoclonal antibody conjugated to horseradish peroxidase for detection.
  • the assay has a limit of detection of 60 pg/mL for plasma and 140 pg/mL for CSF. Results below this concentration were reported as less than reportable (LTR).
  • CSF concentrations of sAPPa and sAPPP were determined using the sAPPot/sAPPp Multi-spot assay (Mesoscale Discovery (Gaithersburg, MD)). CSF was thawed on ice, then diluted 1 : 10 into 1% BSA in TBS-T (10 mM Tris buffer, pH 8.0, 150 mM NaCl, 0.1% Tween-20). The assay was performed as per the manufacturer's protocol. The assay had lower limit of quantification values of 0.05 ng/ml for sAPPa and 0.03 ng/mL for sAPPp.
  • Figures 12A-E summarize the pharmacodynamics behavior of the antibodies.
  • plasma Abetai_ 4 o levels remained unchanged following anti-gD adiminstration, but transiently decreased following anti-TfR/BACEl administration. Both variants reduced plasma Abetai_ 4 o levels, with a maximal inhibition of 50% achieved 1 day post-dosing.
  • Plasma Abetai_4o levels gradually recovered, with animals given anti-TfR'/BACEl returning to baseline Abetai_ 40 levels around 14 days post-dose.
  • Abetai_ 40 levels returned to baseline levels between 21 and 30 days post-dose in animals treated with anti-TfR 2 /BACEl .
  • sAPPa production was stimulated during BACEl inhibition by both anti- TfRVBACEl and anti-TfR 2 /BACEl , and the response correlated inversely with the level of inhibition observed for ⁇ and Abetai_ 4 o.
  • SAPPa and sAPP are the primary processing products of amyloid precursor protein (APP), and their levels are highly correlated.
  • the ratio of sAPPp/sAPPa normalizes the results to potential changes in basal APP expression or potential preanalytical differences in CSF collection and handling over the course of the study.
  • the ratio of CSF sAPPp/sAPPa with anti-TfRVBACEl demonstrated a more robust PD effect than anti-TfR /BACEl .
  • target i.e. BACEl
  • the PD response for anti-TfR 52A /BACEl and anti-TfR 53A /BACEl also correlates with the duration of antibody exposure and a reduced affinity TfR arm shows increased reduction in ⁇ 40 (data not shown). These data also support target engagement by these bispecific antibodies .
  • bispecific antibodies anti-TfRVBACEl or anti-TfR /BACEl, as in the previous example.
  • These bispecific antibodies were in a human IgGl format with D265A and N297G mutations abrogating effector function.
  • an anti-gD molecule on human IgGl was used.
  • a bivalent anti-BACEl antibody which is the same clone used for the bispecific antbibodies.
  • the antibodies were administered by a single intravenous (IV) bolus injection into the saphenous vein at a dose of 30 mg/kg to conscious cynomolgus monkeys with indwelling cisterna magna catheters.
  • Baseline CSF samples were collected 24 and 48 hours prior to dosing, and another CSF sample was collected 24 hours post-dose (as shown schematically in Figure 14).
  • animals were perfused with saline and brains were harvested for analysis of antibody concentrations. Different brain regions were homogenized in 1% NP-40 (Cal-Biochem) in PBS containing Complete Mini EDTA-free protease inhibitor cocktail tablets (Roche Diagnostics).
  • Anti-TfR 2 /BACEl had ⁇ 3-fold improved brain uptake over control IgG (average ⁇ 2 nM), and anti-TfRVBACEl had the best brain uptake, ⁇ 15-fold greater than control IgG (average -10 nM).
  • the brain antibody concentrations for the different antibodies correlated with the pharmacodynamics response seen in CSF in our studies, with anti-TfRVBACEl having the best brain uptake and most robust pharmacodynamics effect, and anti-TfR /BACEl having less brain uptake and a more modest effect.
  • TfR-binding bispecific antibodies improve uptake in brain of non-human primates.
  • primates as in mice, there is likely an optimum affinity to TfR that best balances brain uptake and TfR-mediated clearance.
  • the higher affinity anti-TfRVBACEl demonstrates good brain uptake, and is affected by peripheral target-mediated clearance.
  • TfR 2 /BACEl has improved clearance properties, but appears to have such low binding for TfR as to not be able to be efficiently transported by TfR (much in the same way that the lowest-affinity anti-TfR antibody TfR E in US2012/0171120 passes some affinity threshold beyond which the affinity is too low to permit sufficient interaction between the antibody and TfR such that the antibody would remain associated with TfR as TfR begins the translocation process).
  • mice Pharmacokinetic analysis and reticulocyte count following a single antibody administration in mice were performed as follows. Wild type female C57B/6 mice ages 6-8 weeks were used for all studies. The animals' care was in accordance with institutional guidelines. Mice were dosed intravenously with a single 50mg/kg dose of either an anti-gD antibody (murine IgG2a) with the LALAPG mutations, an anti-TfR D /BACEl antibody
  • IgG2a (allotype a)/ anti-mouse IgG2a (allotype a) ELISA NUNC 384-well Maxisorp iirtmunoplates (Neptune, NJ) were coated with mouse anti-mouse IgG2a allotype A, an allotype A specific antibody (BD/Pharrnigen San Jose, CA), overnight at 4°C. Plates were blocked with PBS, 0.5% BSA for 1 hour at 25°C Each antibody (anti-gD and the anti- TfR/BACEl bispecific variants) was used as a standard to quantify respective antibody concentrations.
  • Bound biotin-conjugated antibody was detected with horseradish peroxidase-conjugated streptavidin (GE Helathcare Life Sciences, Pittsburgh, PA). Samples were developed using 3,3',5,5'- tetramethyl benzidine (TMB) (KPL, Inc., Gaithersburg, MD) and absorbance measured at 450 nm on a Multiskan Ascent reader (Thermo Scientific, Hudson, NH). Concentrations were determined from the standard curve using a four-parameter non-linear regression program. The assay had lower limit of quantification (LLOQ) values of 78.13 ng/ml in plasma. Statistical analysis of differences between experimental groups was performed using a two-tailed unpaired t-test.
  • mice Upon administration of the anti-TfR D /BACEl antibodies containing the Fc LALAPG mutations, the mice displayed no clinical symptoms as had been previously observed using antibodies with full effector function. See Couch et al., Sci. Trans. Med. 5:183ra57 (2013).
  • Figure 20 shows the results of the pharmacokinetic analysis.
  • bispecific variants were made containing mutations in the IgG constant domain and specifically in the Fc eceptor-neonate (FcRn) binding domain (FcRn fflGH mutations).
  • the FcRn binding domain has been implicated in the maternal-fetal transfer of antibodies. See Story et al., J. Exp. Med., 180:2377 2381, 1994.
  • the amino acid substitutions in the FcRn binding domain increase the affinity of the constant domain for the FcRn thereby increasing the half-life of the antibody.
  • FcRn binding domain mutations M252Y, S254T and T256E have been described to increase FcRn binding and thus increase the half- life of antibodies. See U.S. Published Patent Application No. 2003/0190311 and Dall' Acqua et al., J. Biol. Chem.
  • FcRn binding domain mutations N434A and Y436I have been described to also increase FcRn binding. See Yeung et al., J. Immunol. 182: 7663-7671 (2009).
  • the YTE (M252Y/S254T/T256E) and AI (N434/Y436I) mutations were incorporated into both anti-TfR 52A /BACEl and anti-TfR 2 /BACEl bispecific antibodies containing either WT human IgGl or effectorless LALAPG or N297G mutations.
  • FcRn HIGH mutation were made in the anti-gD hlgGl antibody as a control. Mutations were constructed using Kunkel mutatgenesis, antibodies were expressed transiently in CHO cells, and proteins were purified using protein A chromatography followed by size exlucsion chromatography (SEC).
  • Binding of FcRn fflGH variant antibodies to FcRn was measured uing BIAcore.
  • Human and cynomolgus monkey FcRn proteins were expressed in CHO and purified using IgG affinity chromatography. Data were acquired on a BIAcore T200 instrument.
  • a series S sensor chip CM5 GE Healthcare, Cat. BRl 00530
  • EDC and NHS reagents according to the supplier's instructions
  • anti-Fab antibody Human Fab capture kit, GE Health care Bio- science. AB SE-7 184, upsala, Sweden
  • antibodies were first injected at a 10 ⁇ /min flow rate to capture approximately 1000 RU on 3 different flow cells (FC), except for FC1 (reference), and then 2- fold serial dilutions of human FcRn (or Cyno FcRn) in pH6 buffer (0.1M sodium phosphate), from low (1 nM) to high (25 ⁇ ) were injected (flow rate: 30 ⁇ /min) one after the other in the same cycle with no regeneration between injections.
  • Sensograms were recorded and subject to reference and buffer subtraction before evaluating by using BIAcore T200 Evaluation Software (version 2.0). Affinities were determined by analyzing the level of binding at steady state based on a 1 : 1 binding model.
  • Binding affinities for LALAPG, N297G, LALAPG.YTE, and LALAPG.AI variants of anti-TfR 52A /BACE 1 are shown in Table 11 below. The data show that the FcRn mGH variants enhance affinity at endosomal (pH 6) to both human and cyno FcRn.
  • a cassette (human TFRC cDNA, SV40 pA, and frt-PGK-em7-Neo-BGHpA-frt) flanked by short homologies to the mouse Tfrc gene was used to modify a Tfrc C57BL/6J BAC (RP23 BAC library) by recombineering.
  • the human TFRC cDNA cassette was inserted at the endogenous ATG and the remainder of Tfrc exon 2 plus the beginning of intron 2 was deleted.
  • the targeted region in the BAC was then retrieved into pBlight-TK (Warming et al. Molecular and Cellular Biology vol. 26 (18) pp. 6913-22 2006) along with flanking genomic Tfrc
  • sequences as homology arms for ES cell targeting corresponds to (assembly NCBI37/mm9): chr.16:32,610,333-32,613,282 and the 2599 bp 3 ' homology arm corresponds to chr.16:32,613, 320-32, 615,918.
  • the final vector was confirmed by DNA sequencing.
  • the Tfrc/TFRC KI vector was linearized with Notl and C57BL/6N C2 ES cells were targeted using standard methods (G418 positive and gancyclovir negative selection). Positive clones were identified using PCR and taqman analysis, and confirmed by sequencing of the modified locus.
  • the antibodies listed in the table below were administered to huTfR knock- in mice in a single 50mg/kg dose and 24 hours later blood was drawn and reticulocytes, hulgl, N297G
  • ADCC assays were also carried out to confirm the effectorless status of LALAPG, LALAPG/YTE, and LALAPG/AI mutation combinations in a human-derived cell line.
  • human erythroleukemia cell line HEL, ATCC
  • PBMCs from healthy human donors carrying either the F/V158 genotype or the FcyRIIIA V/V158 genotype.
  • the V/V158 genotype was also included in this assay due to the known association with increased NK cell-mediated ADCC activity as well as ability to bind IgG4 antibodies (Bowles and Weiner, 2005: Bruhns et al. 2008). Cells were counted and viability was determined by Vi-CELL (Beckman Coulter; Fullerton, CA) following the manufacturer's instructions.
  • PBMCs were isolated by density gradient centrifugation using Uni-SepTM blood separation tubes (Accurate Chemical & Scientific Corp.; Westbury, NY).
  • Target cells in 50 ⁇ ⁇ of assay medium (RPMI-1640 with 1% BSA and 100 units/mL penicillin and streptomycin) were seeded in a 96-well, round-bottom plate at 4 x 10 4 /well.
  • Serial dilutions of test and control antibodies 50 ⁇ were added to the plates containing the target cells, followed by incubation at 37 C with 5% CO2 for 30 minutes to allow opsonization.
  • the final concentrations of antibodies ranged from 0.0051 to 10,000 ng/mL following 5-fold serial dilutions for a total of 10 data points.
  • the reaction was terminated with 1 M H 3 PO 4 , and absorbance was measured at 490 nm (the background, measured at 650 nm was subtracted for each well) using a SpectraMax Plus microplate reader. Absorbance of wells containing only the target cells served as the control for the background (low control), whereas wells containing target cells lysed with Triton-X100 provided the maximum signal available (high control). Antibody-independent cellular cytotoxicity (AICC) was measured in wells containing target and effector cells without the addition of antibody. The extent of specific ADCC was calculated as follows:
  • ADCC values of sample dilutions were plotted against the antibody concentration, and the dose-response curves were fitted to a four-parameter model using SoftMax Pro.
  • results of the ADCC assay are shown in Figure 23.
  • the effector positive anti-human TfR antibody (anti-TfRVgD IgGl WT) elicited significant ADCC activity on the HEL cells.
  • the anti-TfR 52A /BACEl antibody variants containing LALAPG, LALAPG/YTE, or LALAPG/AI mutations did not display any ADCC activity in HEL cells, similar to the negative control anti-TfR 52A /gD N297G antibody.
PCT/US2014/038847 2013-05-20 2014-05-20 Anti-transferrin receptor antibodies and methods of use WO2014189973A2 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
JP2016515030A JP6612214B2 (ja) 2013-05-20 2014-05-20 抗トランスフェリン受容体抗体及び使用方法
CA2908743A CA2908743A1 (en) 2013-05-20 2014-05-20 Anti-transferrin receptor antibodies and methods of use
PE2020002132A PE20210649A1 (es) 2013-05-20 2014-05-20 Anticuerpos receptores de anti-transferrina y metodos de uso
EP19190228.7A EP3594240B1 (en) 2013-05-20 2014-05-20 Anti-transferrin receptor antibodies and methods of use
IL242088A IL242088B2 (en) 2013-05-20 2014-05-20 Anti-transferrin receptor antibodies and methods of use
EA201592213A EA038367B1 (ru) 2013-05-20 2014-05-20 АНТИТЕЛА ПРОТИВ РЕЦЕПТОРА ТРАНСФЕРРИНА (TfR) И СПОСОБЫ ИХ ИСПОЛЬЗОВАНИЯ
MX2015015970A MX2015015970A (es) 2013-05-20 2014-05-20 Anticuerpos receptores de antitransferina y metodos de uso.
CN201480035703.6A CN105358578B (zh) 2013-05-20 2014-05-20 运铁蛋白受体的抗体及其使用方法
AU2014268726A AU2014268726B2 (en) 2013-05-20 2014-05-20 Anti-transferrin receptor antibodies and methods of use
EP14732721.7A EP2999716A2 (en) 2013-05-20 2014-05-20 Anti-transferrin receptor antibodies and methods of use
KR1020157035600A KR102293064B1 (ko) 2013-05-20 2014-05-20 항-트랜스페린 수용체 항체 및 사용 방법
BR112015029009-4A BR112015029009B1 (pt) 2013-05-20 2014-05-20 Anticorpos, formulação farmacêutica e usos de um anticorpo
EP23210472.9A EP4324480A3 (en) 2013-05-20 2014-05-20 Anti-transferrin receptor antibodies and methods of use
MA38632A MA38632B1 (fr) 2013-05-20 2014-05-20 Anticorps anti-récepteur de transferrine et procédés d'utilisation
SG11201509566RA SG11201509566RA (en) 2013-05-20 2014-05-20 Anti-transferrin receptor antibodies and methods of use
UAA201512536A UA119235C2 (uk) 2013-05-20 2014-05-20 Антитіло анти-рецептор трансферину і способи його застосування
PH12015502565A PH12015502565B1 (en) 2013-05-20 2015-11-12 Anti-transferrin receptor antibodies and methods of use
CR20150618A CR20150618A (es) 2013-05-20 2015-11-19 Anticuerpos receptores de antitransferina y métodos de uso
HK16103512.5A HK1215584A1 (zh) 2013-05-20 2016-03-24 運鐵蛋白受體的抗體及其使用方法
AU2020200355A AU2020200355A1 (en) 2013-05-20 2020-01-17 Anti-transferrin receptor antibodies and methods of use
AU2022201773A AU2022201773A1 (en) 2013-05-20 2022-03-15 Anti-transferrin receptor antibodies and methods of use
IL304243A IL304243A (en) 2013-05-20 2023-07-04 Anti-transferrin receptor antibodies and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361825477P 2013-05-20 2013-05-20
US61/825,477 2013-05-20

Publications (2)

Publication Number Publication Date
WO2014189973A2 true WO2014189973A2 (en) 2014-11-27
WO2014189973A3 WO2014189973A3 (en) 2015-03-05

Family

ID=50983167

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/038847 WO2014189973A2 (en) 2013-05-20 2014-05-20 Anti-transferrin receptor antibodies and methods of use

Country Status (23)

Country Link
US (3) US9708406B2 (ru)
EP (3) EP3594240B1 (ru)
JP (1) JP6612214B2 (ru)
KR (1) KR102293064B1 (ru)
CN (1) CN105358578B (ru)
AR (1) AR096364A1 (ru)
AU (3) AU2014268726B2 (ru)
CA (1) CA2908743A1 (ru)
CL (1) CL2015003406A1 (ru)
CR (1) CR20150618A (ru)
EA (1) EA038367B1 (ru)
HK (1) HK1215584A1 (ru)
HR (1) HRP20240107T1 (ru)
IL (2) IL242088B2 (ru)
MA (1) MA38632B1 (ru)
MX (1) MX2015015970A (ru)
PE (2) PE20210649A1 (ru)
PH (1) PH12015502565B1 (ru)
PL (1) PL3594240T3 (ru)
SG (2) SG10202003997UA (ru)
TW (1) TWI573805B (ru)
UA (1) UA119235C2 (ru)
WO (1) WO2014189973A2 (ru)

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016081643A1 (en) * 2014-11-19 2016-05-26 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
WO2016081640A1 (en) * 2014-11-19 2016-05-26 Genentech, Inc. Anti-transferrin receptor / anti-bace1 multispecific antibodies and methods of use
WO2016179257A2 (en) 2015-05-04 2016-11-10 Cytomx Therapeutics, Inc. Anti-cd71 antibodies, activatable anti-cd71 antibodies, and methods of use thereof
WO2016207240A1 (en) * 2015-06-24 2016-12-29 F. Hoffmann-La Roche Ag Anti-transferrin receptor antibodies with tailored affinity
WO2016208695A1 (ja) 2015-06-24 2016-12-29 Jcrファーマ株式会社 血液脳関門を通過する抗ヒトトランスフェリン受容体抗体
WO2017055542A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific anti-human cd20/human transferrin receptor antibodies and methods of use
US9758591B2 (en) 2012-08-24 2017-09-12 The Regents Of The University Of California Antibodies and vaccines for use in treating ROR1 cancers and inhibiting metastasis
EP3218411A2 (en) * 2014-11-14 2017-09-20 Ossianix Inc. Variable new antigen receptors (vnars) directed against transferrin receptor (tfr) and their use
WO2018152285A1 (en) * 2017-02-17 2018-08-23 Denali Therapeutics Inc. Transferrin receptor transgenic models
WO2018165619A1 (en) 2017-03-09 2018-09-13 Cytomx Therapeutics, Inc. Cd147 antibodies, activatable cd147 antibodies, and methods of making and use thereof
US10143187B2 (en) 2017-02-17 2018-12-04 Denali Therapeutics Inc. Transferrin receptor transgenic models
US10364292B2 (en) 2014-01-06 2019-07-30 Hoffmann-La Roche Inc. Monovalent blood brain barrier shuttle modules
WO2019157224A1 (en) 2018-02-07 2019-08-15 Regeneron Pharmaceuticals, Inc. Methods and compositions for therapeutic protein delivery
US10457717B2 (en) 2017-02-17 2019-10-29 Denali Therapeutics Inc. Engineered polypeptides
US10537633B2 (en) 2016-03-04 2020-01-21 Jn Biosciences Llc Antibodies to TIGIT
CN111148757A (zh) * 2017-08-10 2020-05-12 戴纳立制药公司 工程改造的转铁蛋白受体结合多肽
US10688181B2 (en) 2016-06-27 2020-06-23 The Regents Of The University Of California Cancer treatment combinations
WO2020138487A1 (ja) 2018-12-28 2020-07-02 協和キリン株式会社 TfRに結合するバイスペシフィック抗体
US10759864B2 (en) 2016-12-26 2020-09-01 Jcr Pharmaceuticals Co., Ltd. Anti-human transferrin receptor antibody capable of penetrating blood-brain barrier
US10913800B2 (en) 2018-12-21 2021-02-09 Avidity Biosciences, Inc. Anti-transferrin receptor antibodies and uses thereof
US10941205B2 (en) 2015-10-02 2021-03-09 Hoffmann-La Roche Inc. Bispecific anti-human A-beta/human transferrin receptor antibodies and methods of use
WO2021061867A1 (en) 2019-09-23 2021-04-01 Cytomx Therapeutics, Inc. Anti-cd47 antibodies, activatable anti-cd47 antibodies, and methods of use thereof
WO2021076546A1 (en) * 2019-10-14 2021-04-22 Aro Biotherapeutics Company Cd71 binding fibronectin type iii domains
US11097010B2 (en) 2016-08-06 2021-08-24 Ossianix, Inc. In vivo methods for selecting peptides that cross the blood brain barrier, related compositions and methods of use
US11130815B2 (en) 2015-06-24 2021-09-28 Jcr Pharmaceuticals Co., Ltd. Fusion proteins containing a BDNF and an anti-human transferrin receptor antibody
CN113896797A (zh) * 2017-08-01 2022-01-07 Ab工作室有限公司 双特异性抗体及其用途
US11299534B2 (en) 2016-12-14 2022-04-12 Janssen Biotech, Inc. CD8A-binding fibronectin type III domains
US11345739B2 (en) 2016-12-14 2022-05-31 Janssen Biotech, Inc CD137 binding fibronectin type III domains
US11446387B2 (en) 2020-03-27 2022-09-20 Avidity Biosciences, Inc. Compositions and methods of treating muscle dystrophy
US11447539B2 (en) 2016-12-14 2022-09-20 Janssen Biotech, Inc. PD-L1 binding fibronectin type III domains
WO2022226263A1 (en) * 2021-04-23 2022-10-27 The Trustees Of The University Of Pennsylvania Novel compositions with brain-specific targeting motifs and compositions containing same
US11525137B2 (en) 2020-03-19 2022-12-13 Avidity Biosciences, Inc. Compositions and methods of treating Facioscapulohumeral muscular dystrophy
US11643446B2 (en) 2019-12-23 2023-05-09 Denali Therapeutics Inc. Progranulin variants
US11753455B2 (en) 2018-06-21 2023-09-12 Novo Nordisk A/S Compounds for treatment of obesity
US11781138B2 (en) 2019-10-14 2023-10-10 Aro Biotherapeutics Company FN3 domain-siRNA conjugates and uses thereof
US11795232B2 (en) 2017-02-17 2023-10-24 Denali Therapeutics Inc. Engineered transferrin receptor binding polypeptides
US11820824B2 (en) 2020-06-02 2023-11-21 Arcus Biosciences, Inc. Antibodies to TIGIT
US11827702B2 (en) 2021-09-01 2023-11-28 Biogen Ma Inc. Anti-transferrin receptor antibodies and uses thereof
RU2810756C2 (ru) * 2018-12-28 2023-12-28 Киова Кирин Ко., Лтд. Биспецифические антитела, связывающиеся с tfr
US11866742B2 (en) 2017-10-02 2024-01-09 Denali Therapeutics Inc. Fusion proteins comprising enzyme replacement therapy enzymes
WO2024026488A2 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a modified transferrin receptor locus
WO2024026494A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Viral particles retargeted to transferrin receptor 1
US11912779B2 (en) 2021-09-16 2024-02-27 Avidity Biosciences, Inc. Compositions and methods of treating facioscapulohumeral muscular dystrophy
EP4132589A4 (en) * 2020-04-08 2024-05-08 Aliada Therapeutics Inc COMPOSITIONS AND METHODS FOR DELIVERY OF THE BLOOD-BRAIN BARRIER

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10501545B2 (en) * 2015-06-16 2019-12-10 Genentech, Inc. Anti-CLL-1 antibodies and methods of use
WO2018071913A2 (en) 2016-10-14 2018-04-19 Dana-Farber Cancer Institute, Inc. Modular tetrameric bispecific antibody platform
JP2020505330A (ja) 2017-01-06 2020-02-20 アビディティー バイオサイエンシーズ エルエルシー エクソンスキッピングを誘導する核酸ポリペプチド組成物および方法
GB201711809D0 (en) 2017-07-21 2017-09-06 Governors Of The Univ Of Alberta Antisense oligonucleotide
KR20200033880A (ko) 2017-08-02 2020-03-30 스트레스마크 바이오사이언시즈 인코퍼레이티드 항체 결합 활성 알파 시누클레인
GEP20237565B (en) 2017-11-17 2023-11-10 Merck Sharp & Dohme Llc Antibodies specific for immunoglobulin-like transcript 3 (ilt3) and uses thereof
WO2019113393A1 (en) 2017-12-06 2019-06-13 Avidity Biosciences Llc Compositions and methods of treating muscle atrophy and myotonic dystrophy
BR112020013921A2 (pt) * 2018-01-10 2020-12-01 Denali Therapeutics Inc. polipeptídeos de ligação ao receptor de transferrina e usos destes
SG11202006433PA (en) * 2018-02-05 2020-08-28 Jcr Pharmaceuticals Co Ltd Method for delivering drug to muscle
US20210308273A1 (en) 2018-08-02 2021-10-07 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
US11911484B2 (en) 2018-08-02 2024-02-27 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating myotonic dystrophy
US11168141B2 (en) 2018-08-02 2021-11-09 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
WO2020028831A1 (en) * 2018-08-02 2020-02-06 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating fibrodysplasia ossificans progressiva
KR20210081324A (ko) 2018-08-02 2021-07-01 다인 세라퓨틱스, 인크. 근육 표적화 복합체 및 안면견갑상완 근육 이영양증을 치료하기 위한 그의 용도
WO2020037150A2 (en) 2018-08-16 2020-02-20 Denali Therapeutics Inc. Engineered bispecific proteins
CN109336973B (zh) * 2018-10-11 2021-08-24 中国科学院昆明动物研究所 抗转铁蛋白抗体及其用途
KR20200047937A (ko) 2018-10-26 2020-05-08 재단법인 목암생명과학연구소 Ids를 포함하는 융합 단백질 및 이의 용도
JP2023508596A (ja) 2020-01-02 2023-03-02 エフ. ホフマン-ラ ロシュ アーゲー 脳内の治療用抗体の量を決定するための方法
WO2021142260A1 (en) * 2020-01-10 2021-07-15 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for modulation of acvr1
US11633498B2 (en) 2021-07-09 2023-04-25 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating myotonic dystrophy
US11672872B2 (en) 2021-07-09 2023-06-13 Dyne Therapeutics, Inc. Anti-transferrin receptor antibody and uses thereof
US11771776B2 (en) 2021-07-09 2023-10-03 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating dystrophinopathies
US11638761B2 (en) 2021-07-09 2023-05-02 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating Facioscapulohumeral muscular dystrophy
US11969475B2 (en) 2021-07-09 2024-04-30 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating facioscapulohumeral muscular dystrophy
WO2023287238A1 (ko) * 2021-07-15 2023-01-19 한양대학교 산학협력단 뇌혈관장벽 통과 효율이 향상된 항 인간 트랜스페린 수용체 항체, 및 이를 이용한 다중특이적 항체 및 약학 조성물
US11931421B2 (en) 2022-04-15 2024-03-19 Dyne Therapeutics, Inc. Muscle targeting complexes and formulations for treating myotonic dystrophy
WO2024026471A1 (en) 2022-07-29 2024-02-01 Alector Llc Cd98hc antigen-binding domains and uses therefor
TW202405020A (zh) 2022-07-29 2024-02-01 美商阿列克特有限責任公司 轉鐵蛋白受體抗原結合域及其用途
WO2024036333A2 (en) 2022-08-12 2024-02-15 Epibiologics, Inc. Degradation of egfr using a bispecific binding agent

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012075037A1 (en) * 2010-11-30 2012-06-07 Genentech, Inc. Low affinity blood brain barrier receptor antibodies and uses therefor
WO2013177062A2 (en) * 2012-05-21 2013-11-28 Genentech, Inc. Methods for improving safety of blood-brain barrier transport

Family Cites Families (140)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20559A (en) 1858-06-15 Reducing wheel-tires
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
JPS633262A (ja) * 1986-06-24 1988-01-08 Toshiba Corp 網状赤血球の分析方法及び装置
US5567610A (en) 1986-09-04 1996-10-22 Bioinvent International Ab Method of producing human monoclonal antibodies and kit therefor
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
WO1988007089A1 (en) 1987-03-18 1988-09-22 Medical Research Council Altered antibodies
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
US5606040A (en) 1987-10-30 1997-02-25 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
AU634186B2 (en) 1988-11-11 1993-02-18 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
US5175384A (en) 1988-12-05 1992-12-29 Genpharm International Transgenic mice depleted in mature t-cells and methods for making transgenic mice
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
WO1993010819A1 (en) 1991-11-26 1993-06-10 Alkermes, Inc. Process for the preparation of transferrin receptor specific antibody-neuropharmaceutical or diagnostic agent conjugates
US5672683A (en) 1989-09-07 1997-09-30 Alkermes, Inc. Transferrin neuropharmaceutical agent fusion protein
CA2026147C (en) 1989-10-25 2006-02-07 Ravi J. Chari Cytotoxic agents comprising maytansinoids and their therapeutic use
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5229275A (en) 1990-04-26 1993-07-20 Akzo N.V. In-vitro method for producing antigen-specific human monoclonal antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
EP0564531B1 (en) 1990-12-03 1998-03-25 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
JP4124480B2 (ja) 1991-06-14 2008-07-23 ジェネンテック・インコーポレーテッド 免疫グロブリン変異体
GB9114948D0 (en) 1991-07-11 1991-08-28 Pfizer Ltd Process for preparing sertraline intermediates
MX9204374A (es) 1991-07-25 1993-03-01 Idec Pharma Corp Anticuerpo recombinante y metodo para su produccion.
WO1993006217A1 (en) 1991-09-19 1993-04-01 Genentech, Inc. EXPRESSION IN E. COLI OF ANTIBODY FRAGMENTS HAVING AT LEAST A CYSTEINE PRESENT AS A FREE THIOL, USE FOR THE PRODUCTION OF BIFUNCTIONAL F(ab')2 ANTIBODIES
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
DE69334255D1 (de) 1992-02-06 2009-02-12 Novartis Vaccines & Diagnostic Marker für Krebs und biosynthetisches Bindeprotein dafür
WO1993016177A1 (en) 1992-02-11 1993-08-19 Cell Genesys, Inc. Homogenotization of gene-targeting events
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
ATE191853T1 (de) 1992-07-27 2000-05-15 Us Health Zielgerichte liposome zur blut-hirne schranke
EP0752248B1 (en) 1992-11-13 2000-09-27 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
EP0714409A1 (en) 1993-06-16 1996-06-05 Celltech Therapeutics Limited Antibodies
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US6015555A (en) * 1995-05-19 2000-01-18 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical or diagnostic agent conjugates
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
GB9603256D0 (en) 1996-02-16 1996-04-17 Wellcome Found Antibodies
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
CA2293829C (en) 1997-06-24 2011-06-14 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
AU759779B2 (en) 1997-10-31 2003-05-01 Genentech Inc. Methods and compositions comprising glycoprotein glycoforms
US6610833B1 (en) 1997-11-24 2003-08-26 The Institute For Human Genetics And Biochemistry Monoclonal human natural antibodies
BR9813365A (pt) 1997-12-05 2004-06-15 Scripps Research Inst Método para produção e humanização de um anticorpo monoclonal de rato
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
DK1068241T3 (da) 1998-04-02 2008-02-04 Genentech Inc Antistofvarianter og fragmenter deraf
PT1071700E (pt) 1998-04-20 2010-04-23 Glycart Biotechnology Ag Modificação por glicosilação de anticorpos para melhorar a citotoxicidade celular dependente de anticorpos
ES2694002T3 (es) 1999-01-15 2018-12-17 Genentech, Inc. Polipéptido que comprende una región Fc de IgG1 humana variante
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
PT1914244E (pt) 1999-04-09 2013-07-26 Kyowa Hakko Kirin Co Ltd Processo para regular a actividade de moléculas funcionais sob o ponto de vista imunológico
WO2001025454A2 (en) 1999-10-04 2001-04-12 Medicago Inc. Method for regulating transcription of foreign genes in the presence of nitrogen
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
JP4668498B2 (ja) 1999-10-19 2011-04-13 協和発酵キリン株式会社 ポリペプチドの製造方法
CA2393869A1 (en) 1999-12-15 2001-06-21 Genetech,Inc. Shotgun scanning, a combinatorial method for mapping functional protein epitopes
WO2001049698A1 (en) 1999-12-29 2001-07-12 Immunogen, Inc. Cytotoxic agents comprising modified doxorubicins and daunorubicins and their therapeutic use
CN101670105B (zh) 2000-02-24 2014-08-06 华盛顿大学 螯合淀粉样蛋白β肽的人源化抗体
NZ521540A (en) 2000-04-11 2004-09-24 Genentech Inc Multivalent antibodies and uses therefor
EP1309341A2 (en) 2000-07-07 2003-05-14 Lars Lannfelt Prevention and treatment of alzheimer's disease
CA2785941C (en) 2000-10-06 2017-01-10 Kyowa Hakko Kirin Co., Ltd. Antibody composition-producing cell
US7064191B2 (en) 2000-10-06 2006-06-20 Kyowa Hakko Kogyo Co., Ltd. Process for purifying antibody
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
ATE378403T1 (de) 2000-11-30 2007-11-15 Medarex Inc Transchromosomale transgen-nagetiere zur herstellung von humänen antikörpern
PE20020574A1 (es) 2000-12-06 2002-07-02 Wyeth Corp Anticuerpos humanizados que reconocen el peptido amiloideo beta
ES2649037T3 (es) 2000-12-12 2018-01-09 Medimmune, Llc Moléculas con semividas prolongadas, composiciones y usos de las mismas
CN1555411A (zh) 2001-08-03 2004-12-15 ���迨�����\���ɷݹ�˾ 抗体-依赖性细胞毒性增大的抗体糖基化变体
US20040241164A1 (en) 2001-08-17 2004-12-02 Bales Kelly Renee Use of antibodies having high affinity for soluble ab to treat conditions and diseases related to ass
WO2003016466A2 (en) 2001-08-17 2003-02-27 Eli Lilly And Company ANTI-Aβ ANTIBODIES
EP1443961B1 (en) 2001-10-25 2009-05-06 Genentech, Inc. Glycoprotein compositions
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
MY139983A (en) 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
AU2003236018A1 (en) 2002-04-09 2003-10-20 Kyowa Hakko Kirin Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcGamma RECEPTOR IIIa
JPWO2003085107A1 (ja) 2002-04-09 2005-08-11 協和醗酵工業株式会社 ゲノムが改変された細胞
CA2481925A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Therapeutic agent for patients having human fc.gamma.riiia
EP1498490A4 (en) 2002-04-09 2006-11-29 Kyowa Hakko Kogyo Kk PROCESS FOR PREPARING ANTIBODY COMPOSITION
US7691568B2 (en) 2002-04-09 2010-04-06 Kyowa Hakko Kirin Co., Ltd Antibody composition-containing medicament
JP4628679B2 (ja) 2002-04-09 2011-02-09 協和発酵キリン株式会社 Gdp−フコースの輸送に関与する蛋白質の活性が低下または欠失した細胞
JP4753578B2 (ja) 2002-06-03 2011-08-24 ジェネンテック, インコーポレイテッド 合成抗体ファージライブラリー
ES2347144T3 (es) * 2002-08-30 2010-10-26 Biorexis Pharmaceutical Corporation Proteinas de fusion de la transferrina modificada que comprenden dominos aino o carboxilo de transferrina duplicados.
WO2004029629A1 (en) 2002-09-27 2004-04-08 Janssen Pharmaceutica N.V. N-11 truncated amyloid-beta nomoclonal antibodies, compositions, methods and uses
US20040146512A1 (en) 2002-10-09 2004-07-29 Arnon Rosenthal Methods of treating Alzheimer's disease using antibodies directed against amyloid beta peptide and compositions thereof
US7361740B2 (en) 2002-10-15 2008-04-22 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US20050142141A1 (en) 2002-11-27 2005-06-30 Pardridge William M. Delivery of enzymes to the brain
EP1572744B1 (en) 2002-12-16 2010-06-09 Genentech, Inc. Immunoglobulin variants and uses thereof
WO2004065416A2 (en) 2003-01-16 2004-08-05 Genentech, Inc. Synthetic antibody phage libraries
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
US7871607B2 (en) 2003-03-05 2011-01-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
WO2005028511A2 (en) 2003-03-28 2005-03-31 Centocor, Inc. Anti-amyloid antibodies, compositions, methods and uses
PE20050627A1 (es) 2003-05-30 2005-08-10 Wyeth Corp Anticuerpos humanizados que reconocen el peptido beta amiloideo
US8597911B2 (en) * 2003-06-11 2013-12-03 Chugai Seiyaku Kabushiki Kaisha Process for producing antibodies
WO2005035586A1 (ja) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. 融合蛋白質組成物
EP1705251A4 (en) 2003-10-09 2009-10-28 Kyowa Hakko Kirin Co Ltd PROCESS FOR PRODUCING ANTIBODY COMPOSITION BY RNA INHIBITION OF FUNCTION OF $ G (A) 1,6-FUCOSYLTRANSFERASE
RS55723B1 (sr) 2003-11-05 2017-07-31 Roche Glycart Ag Molekuli koji se vezuju za antigen sa povećanim afinitetom vezivanja za fc receptor i efektornom funkcijom
BR122018071968B8 (pt) 2003-11-06 2021-07-27 Seattle Genetics Inc conjugado de anticorpo-droga, composição farmacêutica, artigo de manufatura e uso de um conjugado de anticorpo-droga
WO2005053742A1 (ja) 2003-12-04 2005-06-16 Kyowa Hakko Kogyo Co., Ltd. 抗体組成物を含有する医薬
RU2386638C2 (ru) 2004-03-31 2010-04-20 Дженентек, Инк. Гуманизированные анти-тфр-бета-антитела
US7785903B2 (en) 2004-04-09 2010-08-31 Genentech, Inc. Variable domain library and uses
CA2885854C (en) 2004-04-13 2017-02-21 F. Hoffmann-La Roche Ag Anti-p-selectin antibodies
PT1740616E (pt) 2004-04-30 2012-03-23 Inst Nat Sante Rech Med Anticorpo anti-rtf
EP1765868B1 (en) * 2004-06-07 2016-04-20 MacroGenics, Inc. Transferrin receptor antibodies
MX2007000998A (es) 2004-07-30 2007-07-11 Rinat Neuroscience Corp Anticuerpos dirigidos peptido beta-amiloide y procedimientos que usan los mismos.
TWI380996B (zh) 2004-09-17 2013-01-01 Hoffmann La Roche 抗ox40l抗體
NZ580115A (en) 2004-09-23 2010-10-29 Genentech Inc Cysteine engineered antibody light chains and conjugates
WO2006039470A2 (en) 2004-09-29 2006-04-13 Centocor, Inc. Anti- amyloid antibodies, compositions, methods and uses
JO3000B1 (ar) 2004-10-20 2016-09-05 Genentech Inc مركبات أجسام مضادة .
US8802820B2 (en) * 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
TW200636066A (en) 2004-12-15 2006-10-16 Elan Pharm Inc Humanized antibodies that recognize beta amyloid peptide
PE20061401A1 (es) 2004-12-15 2006-12-23 Neuralab Ltd ANTICUERPOS Aß PARA MEJORAR LA COGNICION
WO2006066089A1 (en) 2004-12-15 2006-06-22 Neuralab Limited Humanized amyloid beta antibodies for use in improving cognition
ES2259270B1 (es) 2005-03-09 2007-11-01 Consejo Superior De Investigaciones Cientificas Metodo de diagnostico in vitro de la enfermedad de alzheimer mediante un anticuerpo monoclonal.
US8142781B2 (en) * 2005-10-07 2012-03-27 Armagen Technologies, Inc. Fusion proteins for blood-brain barrier delivery
WO2007056441A2 (en) 2005-11-07 2007-05-18 Genentech, Inc. Binding polypeptides with diversified and consensus vh/vl hypervariable sequences
WO2007064919A2 (en) 2005-12-02 2007-06-07 Genentech, Inc. Binding polypeptides with restricted diversity sequences
RU2015111675A (ru) 2005-12-12 2015-08-10 Ац Иммуне Са Специфические в отношении амилоида бета (а бета) 1-42 моноклональные антитела, обладающие терапевтическими свойствами
AU2007249408A1 (en) 2006-05-09 2007-11-22 Genentech, Inc. Binding polypeptides with optimized scaffolds
AR062065A1 (es) 2006-07-14 2008-10-15 Ac Immune Sa Anticuerpo humanizado
US10118970B2 (en) 2006-08-30 2018-11-06 Genentech, Inc. Multispecific antibodies
US20080226635A1 (en) 2006-12-22 2008-09-18 Hans Koll Antibodies against insulin-like growth factor I receptor and uses thereof
CN101245107B (zh) * 2007-02-14 2010-10-13 中国人民解放军军事医学科学院生物工程研究所 抗人转铁蛋白受体人源抗体及其应用
CN100592373C (zh) 2007-05-25 2010-02-24 群康科技(深圳)有限公司 液晶显示面板驱动装置及其驱动方法
MX2009013505A (es) 2007-06-12 2010-01-27 Ac Immune Sa Anticuerpos humanizados para amiloide beta.
KR20160017126A (ko) 2007-06-12 2016-02-15 에이씨 이뮨 에스.에이. 모노클로널 항 베타 아밀로이드 항체
JP2010536907A (ja) 2007-08-31 2010-12-02 ニューリミューン セラピューティクス エイジー 患者にアミロイドーシスおよびタンパク質凝集性障害における特異的免疫応答をもたらす方法
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
EP3590965A1 (en) 2011-03-29 2020-01-08 Roche Glycart AG Antibody fc variants
JP6779876B2 (ja) * 2014-11-19 2020-11-04 ジェネンテック, インコーポレイテッド 抗トランスフェリン受容体抗体及びその使用方法

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012075037A1 (en) * 2010-11-30 2012-06-07 Genentech, Inc. Low affinity blood brain barrier receptor antibodies and uses therefor
WO2013177062A2 (en) * 2012-05-21 2013-11-28 Genentech, Inc. Methods for improving safety of blood-brain barrier transport

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
BIEN-LY NGA ET AL: "Transferrin receptor (TfR) trafficking determines brain uptake of TfR antibody affinity variants", THE JOURNAL OF EXPERIMENTAL MEDICINE, ROCKEFELLER UNIVERSITY PRESS, US, vol. 211, no. 2, 10 February 2014 (2014-02-10), pages 233-244, XP009179491, ISSN: 0022-1007 *
Biocompare: "anti-Transferrin Receptor (p90, CD71) (TFRC) antibody (APC) ABIN238474 from antibodies-online", Biocompare.com, 1 August 2014 (2014-08-01), XP055133157, Retrieved from the Internet: URL:http://www.biocompare.com/9776-Antibodies/4144935-anti-Transferrin-Receptor-p90-CD71-TFRC-antibody-APC/ [retrieved on 2014-08-04] *
COUCH JESSICA A ET AL: "Addressing safety liabilities of TfR bispecific antibodies that cross the blood-brain barrier", SCIENCE / SCIENCE TRANSLATIONAL MEDICINE, WASHINGTON, DC : AAAS, US, vol. 5, no. 183, 1 May 2013 (2013-05-01), XP008163922, ISSN: 1946-6242, DOI: 10.1126/SCITRANSLMED.3005338 *
DANIELS T R ET AL: "The transferrin receptor part I: Biology and targeting with cytotoxic antibodies for the treatment of cancer", CLINICAL IMMUNOLOGY, ACADEMIC PRESS, US, vol. 121, no. 2, 1 November 2006 (2006-11-01), pages 144-158, XP024941315, ISSN: 1521-6616, DOI: 10.1016/J.CLIM.2006.06.010 [retrieved on 2006-11-01] *
HOREJSI V ET AL: "MONOCLONAL ANTIBODIES AGAINST HUMAN LEUKOCYTE ANTIGENS II. ANTIBODIES AGAINST CD45 T200 CD3 T3 CD43 CD10 CALLA TRANSFERRIN RECEPTOR T9 A NOVEL BROADLY EXPRESSED 18-KDA ANTIGEN MEM-43 AND A NOVEL ANTIGEN OF RESTRICTED EXPRESSION MEM-74", FOLIA BIOLOGICA (PRAGUE), vol. 34, no. 1, 1 January 1988 (1988-01-01), pages 23-34, XP009179511, ISSN: 0015-5500 *
LSbio: "CD71 / Transferrin Receptor Mouse anti-Human Monoclonal (APC) (MEM-75) Antibody -LS- C46200 -LSBio", , 1 August 2014 (2014-08-01), XP055133366, Retrieved from the Internet: URL:http://www.lsbio.com/Documents/PDF/Antibodies/46016 [retrieved on 2014-08-05] *
PAUL STEVEN M: "Therapeutic antibodies for brain disorders", SCIENCE / SCIENCE TRANSLATIONAL MEDICINE, WASHINGTON, DC : AAAS, US, vol. 3, no. 84, 25 May 2011 (2011-05-25), page 84ps20, XP009155720, ISSN: 1946-6242 *
POLÁKOVÁ K ET AL: "Characterization of a new monoclonal antibody (TR-19) against human transferrin receptor and its application in topographic study.", NEOPLASMA 1991, vol. 38, no. 1, 1991, pages 21-31, XP009179512, ISSN: 0028-2685 *
RUDIKOFF S ET AL: "Single amino acid substitution altering antigen-binding specificity", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, vol. 79, 1 March 1982 (1982-03-01), pages 1979-1983, XP007901436, ISSN: 0027-8424, DOI: 10.1073/PNAS.79.6.1979 *
TAETLE R ET AL: "MECHANISMS OF GROWTH INHIBITION BY ANTI-TRANSFERRIN RECEPTOR MONOCLONAL ANTIBODIES", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 46, no. 4, PART 01, 1 April 1986 (1986-04-01), pages 1758-1763, XP008061610, ISSN: 0008-5472 *
TRACY R. DANIELS ET AL: "The transferrin receptor and the targeted delivery of therapeutic agents against cancer", BIOCHIMICA ET BIOPHYSICA ACTA (BBA) - GENERAL SUBJECTS, vol. 1820, no. 3, 1 March 2012 (2012-03-01), pages 291-317, XP055133141, ISSN: 0304-4165, DOI: 10.1016/j.bbagen.2011.07.016 *
WHITE S ET AL: "COMBINATIONS OF ANTI-TRANSFERRIN RECEPTOR MONOCLONAL ANTIBODIES INHIBIT HUMAN TUMOR CELL GROWTH IN-VITRO AND IN-VIVO EVIDENCE FOR SYNERGISTIC ANTIPROLIFERATIVE EFFECTS", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 50, no. 19, 1 January 1990 (1990-01-01), pages 6295-6301, XP008073808, ISSN: 0008-5472 *

Cited By (104)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10344096B2 (en) 2012-08-24 2019-07-09 The Regents Of The University Of California Antibodies and vaccines for use in treating ROR1 cancers and inhibiting metastasis
US11312787B2 (en) 2012-08-24 2022-04-26 The Regents Of The University Of California Antibodies and vaccines for use in treating ROR1 cancers and inhibiting metastasis
US9758591B2 (en) 2012-08-24 2017-09-12 The Regents Of The University Of California Antibodies and vaccines for use in treating ROR1 cancers and inhibiting metastasis
US10364292B2 (en) 2014-01-06 2019-07-30 Hoffmann-La Roche Inc. Monovalent blood brain barrier shuttle modules
EP3218411A2 (en) * 2014-11-14 2017-09-20 Ossianix Inc. Variable new antigen receptors (vnars) directed against transferrin receptor (tfr) and their use
EP3218411B1 (en) * 2014-11-14 2022-01-12 Ossianix, Inc. Variable new antigen receptors (vnars) directed against transferrin receptor (tfr) and their use
US11918647B2 (en) 2014-11-14 2024-03-05 Ossianix, Inc. TfR selective binding compounds and related methods
US10722576B2 (en) 2014-11-14 2020-07-28 Ossianix, Inc. TfR selective binding compounds and related methods
US11098129B2 (en) 2014-11-19 2021-08-24 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
WO2016081643A1 (en) * 2014-11-19 2016-05-26 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
US10508151B2 (en) 2014-11-19 2019-12-17 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
US11008403B2 (en) 2014-11-19 2021-05-18 Genentech, Inc. Anti-transferrin receptor / anti-BACE1 multispecific antibodies and methods of use
WO2016081640A1 (en) * 2014-11-19 2016-05-26 Genentech, Inc. Anti-transferrin receptor / anti-bace1 multispecific antibodies and methods of use
US11267896B2 (en) 2015-05-04 2022-03-08 Cytomx Therapeutics, Inc. Anti-CD71 antibodies, activatable anti-CD71 antibodies, and methods of use thereof
WO2016179257A3 (en) * 2015-05-04 2016-12-15 Cytomx Therapeutics, Inc. Anti-cd71 antibodies, activatable anti-cd71 antibodies, and methods of use thereof
WO2016179257A2 (en) 2015-05-04 2016-11-10 Cytomx Therapeutics, Inc. Anti-cd71 antibodies, activatable anti-cd71 antibodies, and methods of use thereof
US10179817B2 (en) 2015-05-04 2019-01-15 Cytomx Therapeutics, Inc. Anti-CD71 antibodies, activatable anti-CD71 antibodies, and methods of use thereof
AU2016258628B2 (en) * 2015-05-04 2021-11-11 Cytomx Therapeutics, Inc Anti-CD71 antibodies, activatable anti-CD71 antibodies, and methods of use thereof
JP2018520991A (ja) * 2015-05-04 2018-08-02 サイトメックス セラピューティクス インコーポレイテッド 抗cd71抗体、活性化可能抗cd71抗体、およびその使用方法
CN108026170B (zh) * 2015-05-04 2022-03-01 西托姆克斯治疗公司 抗cd71抗体、可活化抗cd71抗体及其使用方法
CN114507283A (zh) * 2015-05-04 2022-05-17 西托姆克斯治疗公司 抗cd71抗体、可活化抗cd71抗体及其使用方法
EP4029880A1 (en) 2015-05-04 2022-07-20 CytomX Therapeutics, Inc. Activatable anti-cd71 antibodies, and methods of use thereof
JP2022120070A (ja) * 2015-05-04 2022-08-17 サイトメックス セラピューティクス インコーポレイテッド 抗cd71抗体、活性化可能抗cd71抗体、およびその使用方法
CN108026170A (zh) * 2015-05-04 2018-05-11 西托姆克斯治疗公司 抗cd71抗体、可活化抗cd71抗体及其使用方法
WO2016207240A1 (en) * 2015-06-24 2016-12-29 F. Hoffmann-La Roche Ag Anti-transferrin receptor antibodies with tailored affinity
US11584793B2 (en) 2015-06-24 2023-02-21 Hoffmann-La Roche Inc. Anti-transferrin receptor antibodies with tailored affinity
CN107810199A (zh) * 2015-06-24 2018-03-16 豪夫迈·罗氏有限公司 具有定制亲和力的抗转铁蛋白受体抗体
JP2020097621A (ja) * 2015-06-24 2020-06-25 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft 目的に合わせた親和性を有する抗トランスフェリンレセプター抗体
JP7194796B2 (ja) 2015-06-24 2022-12-22 エフ.ホフマン-ラ ロシュ アーゲー 目的に合わせた親和性を有する抗トランスフェリンレセプター抗体
CN113999312A (zh) * 2015-06-24 2022-02-01 豪夫迈·罗氏有限公司 具有定制亲和力的抗转铁蛋白受体抗体
IL255461B1 (en) * 2015-06-24 2023-06-01 Hoffmann La Roche Antibodies against the transnephrine receptor with adapted affinity
EP4074730A1 (en) * 2015-06-24 2022-10-19 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies with tailored affinity
US11248045B2 (en) 2015-06-24 2022-02-15 Jcr Pharmaceuticals Co., Ltd. Anti-human transferrin receptor antibody permeating blood-brain barrier
TWI792646B (zh) * 2015-06-24 2023-02-11 瑞士商赫孚孟拉羅股份公司 具有特製親和性之抗轉鐵蛋白受體抗體
AU2016282334B2 (en) * 2015-06-24 2022-07-21 F. Hoffmann-La Roche Ag Anti-transferrin receptor antibodies with tailored affinity
WO2016208695A1 (ja) 2015-06-24 2016-12-29 Jcrファーマ株式会社 血液脳関門を通過する抗ヒトトランスフェリン受容体抗体
US10323089B2 (en) 2015-06-24 2019-06-18 Hoffmann-La Roche Inc. Anti-transferrin receptor antibodies with tailored affinity
CN107810199B (zh) * 2015-06-24 2021-11-09 豪夫迈·罗氏有限公司 具有定制亲和力的抗转铁蛋白受体抗体
JP2018520143A (ja) * 2015-06-24 2018-07-26 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft 目的に合わせた親和性を有する抗トランスフェリンレセプター抗体
TWI743038B (zh) * 2015-06-24 2021-10-21 瑞士商赫孚孟拉羅股份公司 具有特製親和性之抗轉鐵蛋白受體抗體
JP2022025120A (ja) * 2015-06-24 2022-02-09 エフ.ホフマン-ラ ロシュ アーゲー 目的に合わせた親和性を有する抗トランスフェリンレセプター抗体
US11958905B2 (en) 2015-06-24 2024-04-16 Jcr Pharmaceuticals Co., Ltd. Fusion proteins containing a BDNF and an anti-human transferrin receptor antibody
US11130815B2 (en) 2015-06-24 2021-09-28 Jcr Pharmaceuticals Co., Ltd. Fusion proteins containing a BDNF and an anti-human transferrin receptor antibody
CN114044829A (zh) * 2015-10-02 2022-02-15 豪夫迈·罗氏有限公司 双特异性抗人A-β/人转铁蛋白受体抗体及使用方法
RU2753390C1 (ru) * 2015-10-02 2021-08-13 Ф. Хоффманн-Ля Рош Аг Биспецифические антитела к человеческому cd20/человеческому рецептору трансферрина и способы их применения
CN114014936A (zh) * 2015-10-02 2022-02-08 豪夫迈·罗氏有限公司 双特异性抗人cd20/人转铁蛋白受体抗体及使用方法
US10941205B2 (en) 2015-10-02 2021-03-09 Hoffmann-La Roche Inc. Bispecific anti-human A-beta/human transferrin receptor antibodies and methods of use
AU2016333512B2 (en) * 2015-10-02 2022-11-17 F. Hoffmann-La Roche Ag Bispecific anti-human CD20/human transferrin receptor antibodies and methods of use
WO2017055542A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific anti-human cd20/human transferrin receptor antibodies and methods of use
US11603411B2 (en) 2015-10-02 2023-03-14 Hoffmann-La Roche Inc. Bispecific anti-human CD20/human transferrin receptor antibodies and methods of use
US10537633B2 (en) 2016-03-04 2020-01-21 Jn Biosciences Llc Antibodies to TIGIT
US11723971B2 (en) 2016-03-04 2023-08-15 JN Biosciences, LLC Antibodies to TIGIT
US10688181B2 (en) 2016-06-27 2020-06-23 The Regents Of The University Of California Cancer treatment combinations
US11654193B2 (en) 2016-06-27 2023-05-23 The Regents Of The University Of California Cancer treatment combinations
US11766482B2 (en) 2016-08-06 2023-09-26 Ossianix, Inc. In vivo methods for selecting peptides that cross the blood brain barrier, related compositions and methods of use
US11097010B2 (en) 2016-08-06 2021-08-24 Ossianix, Inc. In vivo methods for selecting peptides that cross the blood brain barrier, related compositions and methods of use
US11447539B2 (en) 2016-12-14 2022-09-20 Janssen Biotech, Inc. PD-L1 binding fibronectin type III domains
US11299534B2 (en) 2016-12-14 2022-04-12 Janssen Biotech, Inc. CD8A-binding fibronectin type III domains
US11932680B2 (en) 2016-12-14 2024-03-19 Janssen Biotech, Inc. CD8A-binding fibronectin type III domains
US11345739B2 (en) 2016-12-14 2022-05-31 Janssen Biotech, Inc CD137 binding fibronectin type III domains
US11111308B2 (en) 2016-12-26 2021-09-07 Jcr Pharmaceuticals Co., Ltd. Anti-human transferrin receptor antibody capable of penetrating blood-brain barrier
US10759864B2 (en) 2016-12-26 2020-09-01 Jcr Pharmaceuticals Co., Ltd. Anti-human transferrin receptor antibody capable of penetrating blood-brain barrier
US11732023B2 (en) 2017-02-17 2023-08-22 Denali Therapeutics Inc. Engineered polypeptides
WO2018152285A1 (en) * 2017-02-17 2018-08-23 Denali Therapeutics Inc. Transferrin receptor transgenic models
US11795232B2 (en) 2017-02-17 2023-10-24 Denali Therapeutics Inc. Engineered transferrin receptor binding polypeptides
AU2018221704B2 (en) * 2017-02-17 2021-08-19 Denali Therapeutics Inc. Transferrin receptor transgenic models
US11612150B2 (en) 2017-02-17 2023-03-28 Denali Therapeutics Inc. Transferrin receptor transgenic models
US11912778B2 (en) 2017-02-17 2024-02-27 Denali Therapeutics Inc. Methods of engineering transferrin receptor binding polypeptides
US10143187B2 (en) 2017-02-17 2018-12-04 Denali Therapeutics Inc. Transferrin receptor transgenic models
US10457717B2 (en) 2017-02-17 2019-10-29 Denali Therapeutics Inc. Engineered polypeptides
KR102637590B1 (ko) 2017-02-17 2024-02-15 데날리 테라퓨틱스 인크. 트란스페린 수용체 유전자삽입 모델
KR20190135993A (ko) * 2017-02-17 2019-12-09 데날리 테라퓨틱스 인크. 트란스페린 수용체 유전자삽입 모델
WO2018165619A1 (en) 2017-03-09 2018-09-13 Cytomx Therapeutics, Inc. Cd147 antibodies, activatable cd147 antibodies, and methods of making and use thereof
US11566083B2 (en) 2017-08-01 2023-01-31 Ab Studio Inc. Bispecific antibodies and uses thereof
US11440972B2 (en) 2017-08-01 2022-09-13 Ab Studio Inc. Bispecific antibodies and uses thereof
CN113896797A (zh) * 2017-08-01 2022-01-07 Ab工作室有限公司 双特异性抗体及其用途
CN111148757A (zh) * 2017-08-10 2020-05-12 戴纳立制药公司 工程改造的转铁蛋白受体结合多肽
EP3665194A4 (en) * 2017-08-10 2021-07-07 Denali Therapeutics Inc. AFFINITY BASED METHODS FOR USING TRANSFERRIN RECEPTOR BINDING PROTEINS
US11866742B2 (en) 2017-10-02 2024-01-09 Denali Therapeutics Inc. Fusion proteins comprising enzyme replacement therapy enzymes
WO2019157224A1 (en) 2018-02-07 2019-08-15 Regeneron Pharmaceuticals, Inc. Methods and compositions for therapeutic protein delivery
US11753455B2 (en) 2018-06-21 2023-09-12 Novo Nordisk A/S Compounds for treatment of obesity
US11834510B2 (en) 2018-12-21 2023-12-05 Avidity Biosciences, Inc. Anti-transferrin receptor antibodies and uses thereof
US10913800B2 (en) 2018-12-21 2021-02-09 Avidity Biosciences, Inc. Anti-transferrin receptor antibodies and uses thereof
US11028179B2 (en) 2018-12-21 2021-06-08 Avidity Biosciences, Inc. Anti-transferrin receptor antibodies and uses thereof
EP3903816A4 (en) * 2018-12-28 2022-09-07 Kyowa Kirin Co., Ltd. TRANSFERRIN RECEPTOR (TFR) BISPECIFIC ANTIBODY
RU2810756C2 (ru) * 2018-12-28 2023-12-28 Киова Кирин Ко., Лтд. Биспецифические антитела, связывающиеся с tfr
CN113226371A (zh) * 2018-12-28 2021-08-06 协和麒麟株式会社 与TfR结合的双特异性抗体
WO2020138487A1 (ja) 2018-12-28 2020-07-02 協和キリン株式会社 TfRに結合するバイスペシフィック抗体
WO2021061867A1 (en) 2019-09-23 2021-04-01 Cytomx Therapeutics, Inc. Anti-cd47 antibodies, activatable anti-cd47 antibodies, and methods of use thereof
US11628222B2 (en) 2019-10-14 2023-04-18 Aro Biotherapeutics Company CD71 binding fibronectin type III domains
US11781138B2 (en) 2019-10-14 2023-10-10 Aro Biotherapeutics Company FN3 domain-siRNA conjugates and uses thereof
WO2021076546A1 (en) * 2019-10-14 2021-04-22 Aro Biotherapeutics Company Cd71 binding fibronectin type iii domains
US11643446B2 (en) 2019-12-23 2023-05-09 Denali Therapeutics Inc. Progranulin variants
US11555190B2 (en) 2020-03-19 2023-01-17 Avidity Biosciences, Inc. Compositions and methods of treating Facioscapulohumeral muscular dystrophy
US11525137B2 (en) 2020-03-19 2022-12-13 Avidity Biosciences, Inc. Compositions and methods of treating Facioscapulohumeral muscular dystrophy
US11446387B2 (en) 2020-03-27 2022-09-20 Avidity Biosciences, Inc. Compositions and methods of treating muscle dystrophy
US11707532B2 (en) 2020-03-27 2023-07-25 Avidity Biosciences, Inc. Compositions and methods of treating muscle dystrophy
EP4132589A4 (en) * 2020-04-08 2024-05-08 Aliada Therapeutics Inc COMPOSITIONS AND METHODS FOR DELIVERY OF THE BLOOD-BRAIN BARRIER
US11820824B2 (en) 2020-06-02 2023-11-21 Arcus Biosciences, Inc. Antibodies to TIGIT
WO2022226263A1 (en) * 2021-04-23 2022-10-27 The Trustees Of The University Of Pennsylvania Novel compositions with brain-specific targeting motifs and compositions containing same
US11827702B2 (en) 2021-09-01 2023-11-28 Biogen Ma Inc. Anti-transferrin receptor antibodies and uses thereof
US11912779B2 (en) 2021-09-16 2024-02-27 Avidity Biosciences, Inc. Compositions and methods of treating facioscapulohumeral muscular dystrophy
WO2024026488A2 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a modified transferrin receptor locus
WO2024026494A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Viral particles retargeted to transferrin receptor 1

Also Published As

Publication number Publication date
CN105358578A (zh) 2016-02-24
US20180002433A1 (en) 2018-01-04
JP6612214B2 (ja) 2019-11-27
IL242088B1 (en) 2023-08-01
EP4324480A2 (en) 2024-02-21
KR20160011198A (ko) 2016-01-29
US9708406B2 (en) 2017-07-18
AU2014268726A1 (en) 2015-11-19
WO2014189973A3 (en) 2015-03-05
EA038367B1 (ru) 2021-08-16
TWI573805B (zh) 2017-03-11
PH12015502565A1 (en) 2016-02-29
EP2999716A2 (en) 2016-03-30
AU2020200355A1 (en) 2020-02-06
SG10202003997UA (en) 2020-06-29
US20150110791A1 (en) 2015-04-23
US10808036B2 (en) 2020-10-20
SG11201509566RA (en) 2015-12-30
UA119235C2 (uk) 2019-05-27
EP3594240A1 (en) 2020-01-15
CN105358578B (zh) 2021-08-06
CL2015003406A1 (es) 2016-07-15
MX2015015970A (es) 2016-09-08
IL242088B2 (en) 2023-12-01
IL304243A (en) 2023-09-01
PL3594240T3 (pl) 2024-04-02
TW201500373A (zh) 2015-01-01
EP3594240C0 (en) 2023-12-06
IL242088A (ru) 2015-12-31
AR096364A1 (es) 2015-12-23
PE20151926A1 (es) 2016-01-07
MA38632B1 (fr) 2019-10-31
JP2016526878A (ja) 2016-09-08
CR20150618A (es) 2016-02-08
HRP20240107T1 (hr) 2024-04-12
CA2908743A1 (en) 2014-11-27
KR102293064B1 (ko) 2021-08-23
AU2014268726B2 (en) 2019-10-31
AU2022201773A1 (en) 2022-04-07
US20210087288A1 (en) 2021-03-25
EP3594240B1 (en) 2023-12-06
BR112015029009A2 (pt) 2017-09-19
PH12015502565B1 (en) 2016-02-29
MA38632A1 (fr) 2018-09-28
PE20210649A1 (es) 2021-03-26
EA201592213A1 (ru) 2016-04-29
EP4324480A3 (en) 2024-05-08
HK1215584A1 (zh) 2016-09-02

Similar Documents

Publication Publication Date Title
US11098129B2 (en) Anti-transferrin receptor antibodies and methods of use
US20210087288A1 (en) Anti-transferrin receptor antibodies and methods of use
EP3221361B1 (en) Anti-transferrin receptor / anti-bace1 multispecific antibodies and methods of use
EP3313879B1 (en) Anti-transferrin receptor antibodies with tailored affinity
US20180000964A1 (en) Blood brain barrier receptor antibodies and methods of use
US20220033520A1 (en) Modified antibody fcs and methods of use
BR112015029009B1 (pt) Anticorpos, formulação farmacêutica e usos de um anticorpo

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 2908743

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 201480035703.6

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14732721

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2908743

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 242088

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: IDP00201507239

Country of ref document: ID

WWE Wipo information: entry into national phase

Ref document number: 12015502565

Country of ref document: PH

ENP Entry into the national phase

Ref document number: 2016515030

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2014732721

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2014268726

Country of ref document: AU

Date of ref document: 20140520

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15276728

Country of ref document: CO

Ref document number: 002420-2015

Country of ref document: PE

Ref document number: MX/A/2015/015970

Country of ref document: MX

Ref document number: CR2015-000618

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 38632

Country of ref document: MA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015029009

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20157035600

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 201592213

Country of ref document: EA

Ref document number: A201512536

Country of ref document: UA

ENP Entry into the national phase

Ref document number: 112015029009

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20151118